1
|
Nguyen TK, Nguyen ST, Nguyen VT, Na SH, Moon RW, Sattabongkot J, Lau YL, Park WS, Chun WJ, Lu F, Lee SK, Han JH, Han ET. A novel micronemal protein MP38 is involved in the invasion of merozoites into erythrocytes. mBio 2025; 16:e0391724. [PMID: 40202329 PMCID: PMC12077092 DOI: 10.1128/mbio.03917-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 02/21/2025] [Indexed: 04/10/2025] Open
Abstract
The absence of an in vitro cultivation system for Plasmodium vivax hinders the exploration of molecular targets for vaccine development. To address this, functional studies often rely on alternative models, such as P. knowlesi, due to its genetic similarity to P. vivax. This study investigated the role of a novel micronemal protein, PvMP38 (PVX_110945), in both P. vivax and P. knowlesi merozoite invasion of erythrocytes. The full-length ectodomain of PvMP38 was expressed, and polyclonal antibodies were generated to assess its function. PvMP38 was confirmed to localize on micronemal organelle in both P. vivax and P. knowlesi merozoites. In vitro protein-protein interaction assays revealed that PvMP38 binds to Pv12 with high-affinity interaction. A conserved novel complex of Pv12-Pv41-PvMP38 was identified by immunoprecipitation of P. vivax antibodies on P. knowlesi schizont lysates. Linear epitopes of PvMP38 with high and moderate antigenicity were identified in clinical isolates of both species. Invasion inhibition assays demonstrated that a triple antibody combination targeting the PvMP38, Pv12, and Pv41 significantly reduced P. knowlesi merozoite invasion of erythrocytes compared to a single antibody. In addition, CRISPR/Cas9-mediated knockout of P. knowlesi mp38 markedly impaired parasite growth, underscoring its essential role during the asexual stage. These findings identify PvMP38 and its associated complex as promising targets for malaria interventions and highlight the utility of P. knowlesi as a model for investigating P. vivax erythrocyte invasion mechanisms.IMPORTANCEThis manuscript reported an effort in malaria eradication by identifying and functionally characterizing a novel Plasmodium vivax micronemal protein, PvMP38, involved in erythrocyte invasion. A narrow repertoire of an efficacious vaccine targeting P. vivax candidates is being developed due to the lack of continuous in vitro culture. This study addresses a gap in P. vivax research using P. knowlesi as a model for both genome editing and antibody functionality validation. By enhancing the protein-protein interaction screening framework, this study demonstrated that PvMP38 forms a complex with Pv12 and Pv41, opening the approaches to multi-antigen vaccines. The successful application of CRISPR/Cas9 gene editing techniques to disrupt its homolog, the pkmp38 gene, further assesses the protein's significance in the growth and invasion of the parasite. These findings provided valuable insights into the biology of P. vivax and proposed PvMP38 as a promising candidate for malaria intervention strategies.
Collapse
Affiliation(s)
- Tuyet-Kha Nguyen
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon-si, Gangwon-do, South Korea
| | - Sy-Thau Nguyen
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon-si, Gangwon-do, South Korea
- Institue of Clinical Infectious Diseases, 108 Military Central Hospital, Hanoi, Vietnam
| | - Van-Truong Nguyen
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon-si, Gangwon-do, South Korea
| | - Sung-Hun Na
- Department of Obstetrics and Gynecology, Kangwon National University School of Medicine, Chuncheon-si, Gangwon-do, South Korea
| | - Robert W. Moon
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, England, United Kingdom
| | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Yee Ling Lau
- Department of Parasitology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Won-Sun Park
- Department of Physiology, School of Medicine, Kangwon National University, Chuncheon-si, Gangwon-do, South Korea
| | - Wan-Joo Chun
- Department of Pharmacology, School of Medicine, Kangwon National University, Chuncheon-si, Gangwon-do, South Korea
| | - Feng Lu
- Department of Pathogen Biology and Immunology, School of Medicine, Yangzhou University, Yangzhou, Jiangsu, China
| | - Seong-Kyun Lee
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon-si, Gangwon-do, South Korea
| | - Jin-Hee Han
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon-si, Gangwon-do, South Korea
| | - Eun-Taek Han
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon-si, Gangwon-do, South Korea
| |
Collapse
|
2
|
Zhou X, Zhang Q, Chen JH, Dai JF, Kassegne K. Revisiting the antigen markers of vector-borne parasitic diseases identified by immunomics: identification and application to disease control. Expert Rev Proteomics 2024; 21:205-216. [PMID: 38584506 DOI: 10.1080/14789450.2024.2336994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 03/03/2024] [Indexed: 04/09/2024]
Abstract
INTRODUCTION Protein microarray is a promising immunomic approach for identifying biomarkers. Based on our previous study that reviewed parasite antigens and recent parasitic omics research, this article expands to include information on vector-borne parasitic diseases (VBPDs), namely, malaria, schistosomiasis, leishmaniasis, babesiosis, trypanosomiasis, lymphatic filariasis, and onchocerciasis. AREAS COVERED We revisit and systematically summarize antigen markers of vector-borne parasites identified by the immunomic approach and discuss the latest advances in identifying antigens for the rational development of diagnostics and vaccines. The applications and challenges of this approach for VBPD control are also discussed. EXPERT OPINION The immunomic approach has enabled the identification and/or validation of antigen markers for vaccine development, diagnosis, disease surveillance, and treatment. However, this approach presents several challenges, including limited sample size, variability in antigen expression, false-positive results, complexity of omics data, validation and reproducibility, and heterogeneity of diseases. In addition, antigen involvement in host immune evasion and antigen sensitivity/specificity are major issues in its application. Despite these limitations, this approach remains promising for controlling VBPD. Advances in technology and data analysis methods should continue to improve candidate antigen identification, as well as the use of a multiantigen approach in diagnostic and vaccine development for VBPD control.
Collapse
Affiliation(s)
- Xia Zhou
- MOE Key Laboratory of Geriatric Diseases and Immunology, School of Biology & Basic Medical Science, Suzhou Medical College of Soochow University, Suzhou, China
| | - Qianqian Zhang
- Institute of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, China
| | - Jun-Hu Chen
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute of Parasitic Diseases, Chinese Center for Diseases Control and Prevention (Chinese Center for Tropical Diseases Research); National Health Commission of the People's Republic of China (NHC) Key Laboratory of Parasite and Vector Biology; World Health Organization (WHO) Collaborating Center for Tropical Diseases; National Center for International Research on Tropical Diseases, Shanghai, People's Republic of China
- Hainan Tropical Diseases Research Center (Hainan Sub-Center, Chinese Center for Tropical Diseases Research), Haikou, China
| | - Jian-Feng Dai
- Institute of Biology and Medical Sciences, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, China
| | - Kokouvi Kassegne
- School of Global Health, Chinese Center for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- One Health Center, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
3
|
Lee SK, Nguyen TK, Mohring F, Han JH, Firdaus ER, Na SH, Park WS, Moon RW, Han ET. Merozoite surface protein 1 paralog is involved in the human erythrocyte invasion of a zoonotic malaria, Plasmodium knowlesi. Front Cell Infect Microbiol 2023; 13:1314533. [PMID: 38111629 PMCID: PMC10726050 DOI: 10.3389/fcimb.2023.1314533] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 11/15/2023] [Indexed: 12/20/2023] Open
Abstract
The zoonotic malaria parasite Plasmodium knowlesi is an important public health concern in Southeast Asia. Invasion of host erythrocytes is essential for parasite growth, and thus, understanding the repertoire of parasite proteins that enable this process is vital for identifying vaccine candidates and how some species are able to cause zoonotic infection. Merozoite surface protein 1 (MSP1) is found in all malaria parasite species and is perhaps the most well-studied as a potential vaccine candidate. While MSP1 is encoded by a single gene in P. falciparum, all other human infective species (P. vivax, P. knowlesi, P. ovale, and P. malariae) additionally encode a divergent paralogue known as MSP1P, and little is known about its role or potential functional redundancy with MSP1. We, therefore, studied the function of P. knowlesi merozoite surface protein 1 paralog (PkMSP1P), using both recombinant protein and CRISPR-Cas9 genome editing. The recombinant 19-kDa C-terminus of PkMSP1P (PkMSP1P-19) was shown to bind specifically to human reticulocytes. However, immunoblotting data suggested that PkMSP1P-19-induced antibodies can recognize PkMSP1-19 and vice versa, confounding our ability to separate the properties of these two proteins. Targeted disruption of the pkmsp1p gene profoundly impacts parasite growth, demonstrating for the first time that PkMSP1P is important in in vitro growth of P. knowlesi and likely plays a distinct role from PkMSP1. Importantly, the MSP1P KO also enabled functional characterization of the PkMSP1P-19 antibodies, revealing clear immune cross-reactivity between the two paralogues, highlighting the vital importance of genetic studies in contextualizing recombinant protein studies.
Collapse
Affiliation(s)
- Seong-Kyun Lee
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
| | - Tuyet Kha Nguyen
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
| | - Franziska Mohring
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Jin-Hee Han
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
| | - Egy Rahman Firdaus
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
| | - Sung-Hun Na
- Department of Obstetrics and Gynecology, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
| | - Won-Sun Park
- Department of Physiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
| | - Robert W. Moon
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Eun-Taek Han
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
| |
Collapse
|
4
|
Lyons FMT, Gabriela M, Tham WH, Dietrich MH. Plasmodium 6-Cysteine Proteins: Functional Diversity, Transmission-Blocking Antibodies and Structural Scaffolds. Front Cell Infect Microbiol 2022; 12:945924. [PMID: 35899047 PMCID: PMC9309271 DOI: 10.3389/fcimb.2022.945924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 06/22/2022] [Indexed: 11/30/2022] Open
Abstract
The 6-cysteine protein family is one of the most abundant surface antigens that are expressed throughout the Plasmodium falciparum life cycle. Many members of the 6-cysteine family have critical roles in parasite development across the life cycle in parasite transmission, evasion of the host immune response and host cell invasion. The common feature of the family is the 6-cysteine domain, also referred to as s48/45 domain, which is conserved across Aconoidasida. This review summarizes the current approaches for recombinant expression for 6-cysteine proteins, monoclonal antibodies against 6-cysteine proteins that block transmission and the growing collection of crystal structures that provide insights into the functional domains of this protein family.
Collapse
Affiliation(s)
- Frankie M. T. Lyons
- The Walter and Eliza Hall Institute of Medical Research, Infectious Diseases and Immune Defence Division, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Mikha Gabriela
- The Walter and Eliza Hall Institute of Medical Research, Infectious Diseases and Immune Defence Division, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Wai-Hong Tham
- The Walter and Eliza Hall Institute of Medical Research, Infectious Diseases and Immune Defence Division, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Melanie H. Dietrich
- The Walter and Eliza Hall Institute of Medical Research, Infectious Diseases and Immune Defence Division, Parkville, VIC, Australia
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
5
|
Zhang Y, Liu F, Zhao Y, Yang F, Bai J, Jia X, Roobsoong W, Sattabongkot J, Cui L, Cao Y, Luo E, Wang M. Evaluation of two Plasmodium vivax sexual stage antigens as transmission-blocking vaccine candidates. Parasit Vectors 2021; 14:407. [PMID: 34399829 PMCID: PMC8366161 DOI: 10.1186/s13071-021-04909-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 07/30/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Plasmodium vivax transmission-blocking vaccines (TBVs) are receiving increasing attention. Based on excellent transmission-blocking activities of the PbPH (PBANKA_0417200) and PbSOP26 (PBANKA_1457700) antigens in Plasmodium berghei, their orthologs in P. vivax, PVX_098655 (PvPH) and PVX_101120 (PvSOP26), were selected for the evaluation of their potential as TBVs. METHODS Fragments of PvPH (amino acids 22-304) and PvSOP26 (amino acids 30-272) were expressed in the yeast expression system. The recombinant proteins were used to immunize mice to obtain antisera. The transmission-reducing activities of these antisera were evaluated using the direct membrane feeding assay (DMFA) using Anopheles dirus mosquitoes and P. vivax clinical isolates. RESULTS The recombinant proteins PvPH and PvSOP26 induced robust antibody responses in mice. The DMFA showed that the anti-PvSOP26 sera significantly reduced oocyst densities by 92.0 and 84.1% in two parasite isolates, respectively, whereas the anti-PvPH sera did not show evident transmission-reducing activity. The variation in the DMFA results was unlikely due to the genetic polymorphisms of the two genes since their respective sequences were identical in the clinical P. vivax isolates. CONCLUSION PvSOP26 could be a promising TBV candidate for P. vivax, which warrants further evaluation.
Collapse
Affiliation(s)
- Yongzhe Zhang
- Department of Pathogen Biology, College of Basic Medical Sciences, China Medical University, Shenyang, 110122, Liaoning, China
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, China
| | - Fei Liu
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, 110122, Liaoning, China
| | - Yan Zhao
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, 110122, Liaoning, China
| | - Fan Yang
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, 110122, Liaoning, China
| | - Jie Bai
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, 110122, Liaoning, China
| | - Xitong Jia
- Department of Pathogen Biology, College of Basic Medical Sciences, China Medical University, Shenyang, 110122, Liaoning, China
| | - Wanlapa Roobsoong
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Liwang Cui
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, 3720 Spectrum Boulevard, Suite 304, Tampa, FL, 33612-9415, USA
| | - Yaming Cao
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, 110122, Liaoning, China
| | - Enjie Luo
- Department of Pathogen Biology, College of Basic Medical Sciences, China Medical University, Shenyang, 110122, Liaoning, China.
| | - Meilian Wang
- Department of Pathogen Biology, College of Basic Medical Sciences, China Medical University, Shenyang, 110122, Liaoning, China.
| |
Collapse
|
6
|
Hang JW, Tukijan F, Lee EQH, Abdeen SR, Aniweh Y, Malleret B. Zoonotic Malaria: Non- Laverania Plasmodium Biology and Invasion Mechanisms. Pathogens 2021; 10:889. [PMID: 34358039 PMCID: PMC8308728 DOI: 10.3390/pathogens10070889] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/09/2021] [Accepted: 07/09/2021] [Indexed: 12/27/2022] Open
Abstract
Malaria, which is caused by Plasmodium parasites through Anopheles mosquito transmission, remains one of the most life-threatening diseases affecting hundreds of millions of people worldwide every year. Plasmodium vivax, which accounts for the majority of cases of recurring malaria caused by the Plasmodium (non-Laverania) subgenus, is an ancient and continuing zoonosis originating from monkey hosts probably outside Africa. The emergence of other zoonotic malarias (P. knowlesi, P. cynomolgi, and P. simium) further highlights the seriousness of the disease. The severity of this epidemic disease is dependent on many factors, including the parasite characteristics, host-parasite interactions, and the pathology of the infection. Successful infection depends on the ability of the parasite to invade the host; however, little is known about the parasite invasion biology and mechanisms. The lack of this information adds to the challenges to malaria control and elimination, hence enhancing the potential for continuation of this zoonosis. Here, we review the literature describing the characteristics, distribution, and genome details of the parasites, as well as host specificity, host-parasite interactions, and parasite pathology. This information will provide the basis of a greater understanding of the epidemiology and pathogenesis of malaria to support future development of strategies for the control and prevention of this zoonotic infection.
Collapse
Affiliation(s)
- Jing-Wen Hang
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore 117545, Singapore; (J.W.H.); (F.T.); (E.Q.H.L.)
| | - Farhana Tukijan
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore 117545, Singapore; (J.W.H.); (F.T.); (E.Q.H.L.)
| | - Erica-Qian-Hui Lee
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore 117545, Singapore; (J.W.H.); (F.T.); (E.Q.H.L.)
| | - Shifana Raja Abdeen
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore 138648, Singapore;
| | - Yaw Aniweh
- West Africa Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Legon, Accra, Ghana;
| | - Benoit Malleret
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore 117545, Singapore; (J.W.H.); (F.T.); (E.Q.H.L.)
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Biopolis, Singapore 138648, Singapore;
| |
Collapse
|
7
|
Ndegwa DN, Kundu P, Hostetler JB, Marin-Menendez A, Sanderson T, Mwikali K, Verzier LH, Coyle R, Adjalley S, Rayner JC. Using Plasmodium knowlesi as a model for screening Plasmodium vivax blood-stage malaria vaccine targets reveals new candidates. PLoS Pathog 2021; 17:e1008864. [PMID: 34197567 PMCID: PMC8279373 DOI: 10.1371/journal.ppat.1008864] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 07/14/2021] [Accepted: 06/01/2021] [Indexed: 11/18/2022] Open
Abstract
Plasmodium vivax is responsible for the majority of malaria cases outside Africa. Unlike P. falciparum, the P. vivax life-cycle includes a dormant liver stage, the hypnozoite, which can cause infection in the absence of mosquito transmission. An effective vaccine against P. vivax blood stages would limit symptoms and pathology from such recurrent infections, and therefore could play a critical role in the control of this species. Vaccine development in P. vivax, however, lags considerably behind P. falciparum, which has many identified targets with several having transitioned to Phase II testing. By contrast only one P. vivax blood-stage vaccine candidate based on the Duffy Binding Protein (PvDBP), has reached Phase Ia, in large part because the lack of a continuous in vitro culture system for P. vivax limits systematic screening of new candidates. We used the close phylogenetic relationship between P. vivax and P. knowlesi, for which an in vitro culture system in human erythrocytes exists, to test the scalability of systematic reverse vaccinology to identify and prioritise P. vivax blood-stage targets. A panel of P. vivax proteins predicted to function in erythrocyte invasion were expressed as full-length recombinant ectodomains in a mammalian expression system. Eight of these antigens were used to generate polyclonal antibodies, which were screened for their ability to recognize orthologous proteins in P. knowlesi. These antibodies were then tested for inhibition of growth and invasion of both wild type P. knowlesi and chimeric P. knowlesi lines modified using CRISPR/Cas9 to exchange P. knowlesi genes with their P. vivax orthologues. Candidates that induced antibodies that inhibited invasion to a similar level as PvDBP were identified, confirming the utility of P. knowlesi as a model for P. vivax vaccine development and prioritizing antigens for further follow up.
Collapse
Affiliation(s)
- Duncan N. Ndegwa
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
- Department of Biological Sciences, University of Embu, Embu, Kenya
| | - Prasun Kundu
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Hills Road, Cambridge, United Kingdom
| | - Jessica B. Hostetler
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | | | - Theo Sanderson
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Kioko Mwikali
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Lisa H. Verzier
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Rachael Coyle
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Sophie Adjalley
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Julian C. Rayner
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge Biomedical Campus, Hills Road, Cambridge, United Kingdom
| |
Collapse
|
8
|
A novel platform for peptide-mediated affinity capture and LC-MS/MS identification of host receptors involved in Plasmodium invasion. J Proteomics 2020; 231:104002. [PMID: 33045431 DOI: 10.1016/j.jprot.2020.104002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 03/12/2020] [Accepted: 09/30/2020] [Indexed: 11/21/2022]
Abstract
Successful Plasmodium falciparum invasion of red blood cells includes the orderly execution of highly specific receptor-ligand molecular interactions between the parasite's proteins and the red blood cell membrane proteins. There is a growing need for elucidating receptor-ligand pairings, which will help in understanding the parasite's biology and provide the fundamental basis for developing prophylactic or therapeutic alternatives leading to mitigating or eliminating this type of malaria. We have thus used Plasmodium falciparum RH5 - derived peptides and ghost red blood cell proteins in synthetic peptide affinity capture assays to identify important host receptors used by Plasmodium spp. in the invasion of red blood cells. LC-MS/MS analysis confirmed the extensively described interaction between PfRH5 and the basigin receptor on the red blood cell membrane. As shown here, tagged synthetic peptides displaying high binding ability to erythrocytes can be used to identify receptors present in protein extracts from ghost red blood cells via affinity capture and LC-MS/MS. SIGNIFICANCE: The article describes a novel approach for identifying red blood cell receptors based on the ability of synthetic peptides having high red blood cell binding capacity to capture Plasmodium spp. receptors on proteins extracted from ghost red blood cells. Specifically, novel methods to identify Plasmodium falciparum reticulocyte binding protein homolog 5 PfRH5 and basigin interaction using a combination of affinity capture and LC-MS/MS assays is described. Identification of these host RBC receptors interacting with malarial parasite proteins is of utmost importance in studying the disease's pathogenesis and will provide crucial information in understanding the parasite's biology. In addition, data from these studies can be used to identify potential therapeutic target(s) to mitigate or eliminate this debilitating disease.
Collapse
|
9
|
Muh F, Kim N, Nyunt MH, Firdaus ER, Han JH, Hoque MR, Lee SK, Park JH, Moon RW, Lau YL, Kaneko O, Han ET. Cross-species reactivity of antibodies against Plasmodium vivax blood-stage antigens to Plasmodium knowlesi. PLoS Negl Trop Dis 2020; 14:e0008323. [PMID: 32559186 PMCID: PMC7304578 DOI: 10.1371/journal.pntd.0008323] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 04/24/2020] [Indexed: 12/22/2022] Open
Abstract
Malaria is caused by multiple different species of protozoan parasites, and interventions in the pre-elimination phase can lead to drastic changes in the proportion of each species causing malaria. In endemic areas, cross-reactivity may play an important role in the protection and blocking transmission. Thus, successful control of one species could lead to an increase in other parasite species. A few studies have reported cross-reactivity producing cross-immunity, but the extent of cross-reactive, particularly between closely related species, is poorly understood. P. vivax and P. knowlesi are particularly closely related species causing malaria infections in SE Asia, and whilst P. vivax cases are in decline, zoonotic P. knowlesi infections are rising in some areas. In this study, the cross-species reactivity and growth inhibition activity of P. vivax blood-stage antigen-specific antibodies against P. knowlesi parasites were investigated. Bioinformatics analysis, immunofluorescence assay, western blotting, protein microarray, and growth inhibition assay were performed to investigate the cross-reactivity. P. vivax blood-stage antigen-specific antibodies recognized the molecules located on the surface or released from apical organelles of P. knowlesi merozoites. Recombinant P. vivax and P. knowlesi proteins were also recognized by P. knowlesi- and P. vivax-infected patient antibodies, respectively. Immunoglobulin G against P. vivax antigens from both immune animals and human malaria patients inhibited the erythrocyte invasion by P. knowlesi. This study demonstrates that there is extensive cross-reactivity between antibodies against P. vivax to P. knowlesi in the blood stage, and these antibodies can potently inhibit in vitro invasion, highlighting the potential cross-protective immunity in endemic areas. In recent years, malaria initiatives have increasingly shifted focus from achieving malaria control to achieving malaria elimination. However, the interventions used are leading to drastic changes in the proportions of different Plasmodium species causing clinical infection, particularly within Southeast Asia. Little is known about how these different parasite species interact/compete in nature or whether exposure to one species could cause some level of protection against another. We examined cross-reactive antibody responses to key parasite proteins with roles in red blood cell invasion and identified novel cross-species reactivity among the closest of malaria affecting the human population (P. vivax and P. knowlesi). This comprehensive analysis provides evidence that cross-reactive immunity could play an important role in areas where species distributions are perturbed by malaria control measures, and future efforts to identify the specific cross-reactive epitopes involved would be invaluable both to our understanding of malaria immunity and vaccine development.
Collapse
Affiliation(s)
- Fauzi Muh
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
| | - Namhyeok Kim
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
| | | | - Egy Rahman Firdaus
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
| | - Jin-Hee Han
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
| | - Mohammad Rafiul Hoque
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
| | - Seong-Kyun Lee
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
| | - Ji-Hoon Park
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
| | - Robert W. Moon
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Yee Ling Lau
- Department of Parasitology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Osamu Kaneko
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Sakamoto, Nagasaki, Japan
| | - Eun-Taek Han
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
- * E-mail:
| |
Collapse
|
10
|
Kassegne K, Abe EM, Cui YB, Chen SB, Xu B, Deng WP, Shen HM, Wang Y, Chen JH, Zhou XN. Contribution of Plasmodium immunomics: potential impact for serological testing and surveillance of malaria. Expert Rev Proteomics 2018; 16:117-129. [PMID: 30513025 DOI: 10.1080/14789450.2019.1554441] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Introduction: Plasmodium vivax (Pv) and P. knowlesi account together for a considerable share of the global burden of malaria, along with P. falciparum (Pf). However, inaccurate diagnosis and undetectable asymptomatic/submicroscopic malaria infections remain very challenging. Blood-stage antigens involved in either invasion of red blood cells or sequestration/cytoadherence of parasitized erythrocytes have been immunomics-characterized, and are vital for the detection of malaria incidence. Areas covered: We review the recent advances in Plasmodium immunomics to discuss serological markers with potential for specific and sensitive diagnosis of malaria. Insights on alternative use of immunomics to assess malaria prevalence are also highlighted. Finally, we provide practical applications of serological markers as diagnostics, with an emphasis on dot immunogold filtration assay which holds promise for malaria diagnosis and epidemiological surveys. Expert commentary: The approach largely contributes to Pf and Pv research in identifying promising non-orthologous antigens able to detect malaria incidence and to differentiate between past and recent infections. However, further studies to profiling naturally acquired immune responses are expected in order to help discover/validate serological markers of no cross-seroreactivity and guide control interventions. More so, the application of immunomics to knowlesi infections would help validate the recently identified antigens and contribute to the discovery of additional biomarkers of exposure, immunity, or both.
Collapse
Affiliation(s)
- Kokouvi Kassegne
- a National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Key Laboratory of Parasite and Vector Biology of the Chinese Ministry of Health , National Centre for International Research on Tropical Diseases, WHO Collaborating Center for Tropical Diseases, Shanghai, People's Republic of China
| | - Eniola Michael Abe
- a National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Key Laboratory of Parasite and Vector Biology of the Chinese Ministry of Health , National Centre for International Research on Tropical Diseases, WHO Collaborating Center for Tropical Diseases, Shanghai, People's Republic of China
| | - Yan-Bing Cui
- a National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Key Laboratory of Parasite and Vector Biology of the Chinese Ministry of Health , National Centre for International Research on Tropical Diseases, WHO Collaborating Center for Tropical Diseases, Shanghai, People's Republic of China
| | - Shen-Bo Chen
- a National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Key Laboratory of Parasite and Vector Biology of the Chinese Ministry of Health , National Centre for International Research on Tropical Diseases, WHO Collaborating Center for Tropical Diseases, Shanghai, People's Republic of China
| | - Bin Xu
- a National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Key Laboratory of Parasite and Vector Biology of the Chinese Ministry of Health , National Centre for International Research on Tropical Diseases, WHO Collaborating Center for Tropical Diseases, Shanghai, People's Republic of China
| | - Wang-Ping Deng
- a National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Key Laboratory of Parasite and Vector Biology of the Chinese Ministry of Health , National Centre for International Research on Tropical Diseases, WHO Collaborating Center for Tropical Diseases, Shanghai, People's Republic of China
| | - Hai-Mo Shen
- a National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Key Laboratory of Parasite and Vector Biology of the Chinese Ministry of Health , National Centre for International Research on Tropical Diseases, WHO Collaborating Center for Tropical Diseases, Shanghai, People's Republic of China
| | - Yue Wang
- b Institute of Parasitic Diseases , Zhejiang Academy of Medical Sciences , Hangzhou , People's Republic of China
| | - Jun-Hu Chen
- a National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Key Laboratory of Parasite and Vector Biology of the Chinese Ministry of Health , National Centre for International Research on Tropical Diseases, WHO Collaborating Center for Tropical Diseases, Shanghai, People's Republic of China
| | - Xiao-Nong Zhou
- a National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Key Laboratory of Parasite and Vector Biology of the Chinese Ministry of Health , National Centre for International Research on Tropical Diseases, WHO Collaborating Center for Tropical Diseases, Shanghai, People's Republic of China
| |
Collapse
|
11
|
Yepes-Pérez Y, López C, Suárez CF, Patarroyo MA. Plasmodium vivax Pv12 B-cell epitopes and HLA-DRβ1*-dependent T-cell epitopes in vitro antigenicity. PLoS One 2018; 13:e0203715. [PMID: 30199554 PMCID: PMC6130872 DOI: 10.1371/journal.pone.0203715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 08/24/2018] [Indexed: 11/18/2022] Open
Abstract
Malaria is an infectious disease caused by parasites from the genus Plasmodium (P. falciparum and P. vivax are responsible for 90% of all clinical cases); it is widely distributed throughout the world’s tropical and subtropical regions. The P. vivax Pv12 protein is involved in invasion, is expressed on merozoite surface and has been recognised by antibodies from individuals exposed to the disease. In this study, B- and T-cell epitopes from Pv12 were predicted and characterised to advance in the design of a peptide-based vaccine against malaria. For evaluating the humoral response of individuals exposed to natural P. vivax infection from two endemic areas in Colombia, BepiPred-1.0 software was used for selecting B-cell epitopes. B-cell epitope 39038 displayed the greatest recognition by naturally-acquired antibodies and induced an IgG2/IgG4 response. NetMHCIIpan-3.1 prediction software was used for selecting peptides having high affinity binding for HLA-DRβ1* allele lineages and this was confirmed by in-vitro binding assays. T-epitopes 39113 and 39117 triggered a memory T-cell response (Stimulation Index≥2) and significant cytokine production. Combining in-silico, in-vitro and functional assays, two Pv12 protein regions (containing peptides 39038, 39040, 39113 and 39117) have thus been characterised as promising vaccine candidates against P. vivax malaria.
Collapse
Affiliation(s)
- Yoelis Yepes-Pérez
- Molecular Biology and Immunology Department, Fundación Instituto de Immunología de Colombia (FIDIC), Bogotá D.C., Colombia
- MSc Programme in Microbiology, Universidad Nacional de Colombia, Bogotá D.C., Colombia
| | - Carolina López
- Molecular Biology and Immunology Department, Fundación Instituto de Immunología de Colombia (FIDIC), Bogotá D.C., Colombia
- PhD Programme in Biomedical and Biological Sciences, Universidad del Rosario, Bogotá D.C., Colombia
| | - Carlos Fernando Suárez
- Bio-mathematics Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá D.C., Colombia
- Universidad de Ciencias Aplicadas y Ambientales (U.D.C.A), Bogotá D.C., Colombia
| | - Manuel Alfonso Patarroyo
- Molecular Biology and Immunology Department, Fundación Instituto de Immunología de Colombia (FIDIC), Bogotá D.C., Colombia
- Basic Sciences Department, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá D.C., Colombia
- * E-mail:
| |
Collapse
|
12
|
Bermúdez M, Moreno-Pérez DA, Arévalo-Pinzón G, Curtidor H, Patarroyo MA. Plasmodium vivax in vitro continuous culture: the spoke in the wheel. Malar J 2018; 17:301. [PMID: 30126427 PMCID: PMC6102941 DOI: 10.1186/s12936-018-2456-5] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 08/13/2018] [Indexed: 01/01/2023] Open
Abstract
Understanding the life cycle of Plasmodium vivax is fundamental for developing strategies aimed at controlling and eliminating this parasitic species. Although advances in omic sciences and high-throughput techniques in recent years have enabled the identification and characterization of proteins which might be participating in P. vivax invasion of target cells, exclusive parasite tropism for invading reticulocytes has become the main obstacle in maintaining a continuous culture for this species. Such advance that would help in defining each parasite protein’s function in the complex process of P. vivax invasion, in addition to evaluating new therapeutic agents, is still a dream. Advances related to maintenance, culture medium supplements and the use of different sources of reticulocytes and parasites (strains and isolates) have been made regarding the development of an in vitro culture for P. vivax; however, only some cultures having few replication cycles have been obtained to date, meaning that this parasite’s maintenance goes beyond the technical components involved. Although it is still not yet clear which molecular mechanisms P. vivax prefers for invading young CD71+ reticulocytes [early maturation stages (I–II–III)], changes related to membrane proteins remodelling of such cells could form part of the explanation. The most relevant aspects regarding P. vivax in vitro culture and host cell characteristics have been analysed in this review to explain possible reasons why the species’ continuous in vitro culture is so difficult to standardize. Some alternatives for P. vivax in vitro culture have also been described.
Collapse
Affiliation(s)
- Maritza Bermúdez
- Receptor-ligand Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Carrera 50 No. 26-20, Bogotá, Colombia
| | - Darwin Andrés Moreno-Pérez
- Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Carrera 50 No. 26-20, Bogotá, Colombia.,Livestock Sciences Faculty, Universidad de Ciencias Aplicadas y Ambientales (U.D.C.A), Calle 222 No. 55-37, Bogotá, DC, Colombia
| | - Gabriela Arévalo-Pinzón
- Receptor-ligand Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Carrera 50 No. 26-20, Bogotá, Colombia
| | - Hernando Curtidor
- Receptor-ligand Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Carrera 50 No. 26-20, Bogotá, Colombia.,Basic Sciences Department, School of Medicine and Health Sciences, Universidad del Rosario, Carrera 24 No. 63C-69, Bogotá, DC, Colombia
| | - Manuel Alfonso Patarroyo
- Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Carrera 50 No. 26-20, Bogotá, Colombia. .,Basic Sciences Department, School of Medicine and Health Sciences, Universidad del Rosario, Carrera 24 No. 63C-69, Bogotá, DC, Colombia.
| |
Collapse
|
13
|
Muh F, Han JH, Nyunt MH, Lee SK, Jeon HY, Ha KS, Park WS, Hong SH, Ahmed MA, Na S, Takashima E, Tsuboi T, Han ET. Identification of a novel merozoite surface antigen of Plasmodium vivax, PvMSA180. Malar J 2017; 16:133. [PMID: 28351409 PMCID: PMC5369000 DOI: 10.1186/s12936-017-1760-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 02/28/2017] [Indexed: 02/06/2023] Open
Abstract
Background Although a number of Plasmodium vivax proteins have been identified, few have been investigated as potential vaccine candidates. This study characterized the Plasmodium vivax merozoite surface antigen 180 (PvMSA180, PVX_094920), a novel P. vivax antigenic protein. Methods The target gene was amplified as four overlapping domains (D1, D2, D3 and D4) to enable expression of the recombinant protein using cell-free and bacterial expression systems. The recombinant PvMSA180 proteins were used in protein microarrays to evaluate the humoral immune response of 72 vivax-infected patients and 24 vivax-naïve individuals. Antibodies produced in mice against the PvMSA180-D1 and -D4 domains were used to assess the subcellular localization of schizont-stage parasites with immunofluorescence assays. A total of 51 pvmsa180 sequences from 12 countries (41 sequences from PlasmoDB and 6 generated in this study) were used to determine the genetic diversity and genealogical relationships with DNAsp and NETWORK software packages, respectively. Results PvMSA180 consists of 1603 amino acids with a predicted molecular mass of 182 kDa, and has a signal peptide at the amino-terminus. A total of 70.8% of patients (51/72) showed a specific antibody response to at least one of the PvMSA180 domains, and 20.8% (15/72) exhibited a robust antibody response to at least three of the domains. These findings suggest that PvMSA180 is targeted by the humoral immune response during natural infection with P. vivax. Immunofluorescence analysis demonstrated that PvMSA180 is localized on the merozoite surface of schizont-stage parasites, and pvmsa180 sequences originating from various geographic regions worldwide showed low genetic diversity. Twenty-two haplotypes were found, and haplotype 6 (Hap_6, 77%) of pvmsa180 was detected in isolates from six countries. Conclusions A novel P. vivax surface protein, PvMSA180, was characterized in this study. Most of P. vivax-infected patients had specific antibodies against particular antigenic domains, indicating that this protein is immunogenic in naturally exposed populations. Genetic analysis of worldwide isolates showed that pvmsa180 is less polymorphic than other well-known candidates and that some haplotypes are common to several countries. However, additional studies with a larger sample size are necessary to evaluate the antibody responses in geographically separated populations, and to identify the function of PvMSA180 during parasite invasion. Electronic supplementary material The online version of this article (doi:10.1186/s12936-017-1760-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Fauzi Muh
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
| | - Jin-Hee Han
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
| | - Myat Htut Nyunt
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea.,Department of Medical Research, Yangon, Republic of the Union of Myanmar
| | - Seong-Kyun Lee
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
| | - Hye-Yoon Jeon
- Department of Cellular and Molecular Biology, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
| | - Kwon-Soo Ha
- Department of Cellular and Molecular Biology, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
| | - Won Sun Park
- Department of Physiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
| | - Seok-Ho Hong
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
| | - Md Atique Ahmed
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
| | - Sunghun Na
- Department of Obstetrics and Gynecology, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
| | - Eizo Takashima
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Ehime, 790-8577, Japan
| | - Takafumi Tsuboi
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Ehime, 790-8577, Japan
| | - Eun-Taek Han
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea.
| |
Collapse
|
14
|
The s48/45 six-cysteine proteins: mediators of interaction throughout the Plasmodium life cycle. Int J Parasitol 2016; 47:409-423. [PMID: 27899328 DOI: 10.1016/j.ijpara.2016.10.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 10/01/2016] [Accepted: 10/05/2016] [Indexed: 01/05/2023]
Abstract
During their life cycle Plasmodium parasites rely upon an arsenal of proteins that establish key interactions with the host and vector, and between the parasite sexual stages, with the purpose of ensuring infection, reproduction and proliferation. Among these is a group of secreted or membrane-anchored proteins known as the six-cysteine (6-cys) family. This is a small but important family with only 14 members thus far identified, each stage-specifically expressed during the parasite life cycle. 6-cys proteins often localise at the parasite surface or interface with the host and vector, and are conserved in different Plasmodium species. The unifying feature of the family is the s48/45 domain, presumably involved in adhesion and structurally related to Ephrins, the ligands of Eph receptors. The most prominent s48/45 members are currently under functional investigation and are being pursued as vaccine candidates. In this review, we examine what is known about the 6-cys family, their structure and function, and discuss future research directions.
Collapse
|
15
|
Kassegne K, Abe EM, Chen JH, Zhou XN. Immunomic approaches for antigen discovery of human parasites. Expert Rev Proteomics 2016; 13:1091-1101. [DOI: 10.1080/14789450.2016.1252675] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- Kokouvi Kassegne
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, WHO Collaborating Centre for Tropical Diseases, National Center for International Research on Tropical Diseases, Key Laboratory of Parasite and Vector Biology of the Chinese Ministry of Health, Shanghai, People’s Republic of China
| | - Eniola Michael Abe
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, WHO Collaborating Centre for Tropical Diseases, National Center for International Research on Tropical Diseases, Key Laboratory of Parasite and Vector Biology of the Chinese Ministry of Health, Shanghai, People’s Republic of China
| | - Jun-Hu Chen
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, WHO Collaborating Centre for Tropical Diseases, National Center for International Research on Tropical Diseases, Key Laboratory of Parasite and Vector Biology of the Chinese Ministry of Health, Shanghai, People’s Republic of China
| | - Xiao-Nong Zhou
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, WHO Collaborating Centre for Tropical Diseases, National Center for International Research on Tropical Diseases, Key Laboratory of Parasite and Vector Biology of the Chinese Ministry of Health, Shanghai, People’s Republic of China
| |
Collapse
|
16
|
Cheng Y, Lu F, Wang B, Li J, Han JH, Ito D, Kong DH, Jiang L, Wu J, Ha KS, Takashima E, Sattabongkot J, Cao J, Nyunt MH, Kyaw MP, Desai SA, Miller LH, Tsuboi T, Han ET. Plasmodium vivax GPI-anchored micronemal antigen (PvGAMA) binds human erythrocytes independent of Duffy antigen status. Sci Rep 2016; 6:35581. [PMID: 27759110 PMCID: PMC5069673 DOI: 10.1038/srep35581] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 10/04/2016] [Indexed: 11/16/2022] Open
Abstract
Plasmodium vivax, a major agent of malaria in both temperate and tropical climates, has been thought to be unable to infect humans lacking the Duffy (Fy) blood group antigen because this receptor is critical for erythrocyte invasion. Recent surveys in various endemic regions, however, have reported P. vivax infections in Duffy-negative individuals, suggesting that the parasite may utilize alternative receptor-ligand pairs to complete the erythrocyte invasion. Here, we identified and characterized a novel parasite ligand, Plasmodium vivax GPI-anchored micronemal antigen (PvGAMA), that bound human erythrocytes regardless of Duffy antigen status. PvGAMA was localized at the microneme in the mature schizont-stage parasites. The antibodies against PvGAMA fragments inhibited PvGAMA binding to erythrocytes in a dose-dependent manner. The erythrocyte-specific binding activities of PvGAMA were significantly reduced by chymotrypsin treatment. Thus, PvGAMA may be an adhesion molecule for the invasion of Duffy-positive and -negative human erythrocytes.
Collapse
Affiliation(s)
- Yang Cheng
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do 200-701, Republic of Korea.,Department of Parasitology, Wuxi Medical School, Jiangnan University, Wuxi, Jiangsu 214122, China.,Laboratory of Malaria and Vector Research (LMVR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, MD 20852, USA
| | - Feng Lu
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do 200-701, Republic of Korea.,Jiangsu Institute of Parasitic Diseases, Wuxi, Jiangsu, People's China
| | - Bo Wang
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do 200-701, Republic of Korea.,Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Jian Li
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do 200-701, Republic of Korea.,Department of Parasitology, College of Basic Medicine, Hubei University of Medicine, Shiyan, Hubei, China
| | - Jin-Hee Han
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do 200-701, Republic of Korea
| | - Daisuke Ito
- Laboratory of Malaria and Vector Research (LMVR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, MD 20852, USA.,Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Ehime 790-8577, Japan
| | - Deok-Hoon Kong
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do 200-701, Republic of Korea
| | - Lubin Jiang
- Key Laboratory of Molecular Virology and Immunology, Unit of Human Parasite Molecular and Cell Biology, Institut Pasteur of Shanghai, Shanghai 200031, China
| | - Jian Wu
- Laboratory of Malaria and Vector Research (LMVR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, MD 20852, USA
| | - Kwon-Soo Ha
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do 200-701, Republic of Korea
| | - Eizo Takashima
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Ehime 790-8577, Japan
| | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Jun Cao
- Jiangsu Institute of Parasitic Diseases, Wuxi, Jiangsu, People's China
| | - Myat Htut Nyunt
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do 200-701, Republic of Korea.,Department of Medical Research, Yangon, Myanmar
| | | | - Sanjay A Desai
- Laboratory of Malaria and Vector Research (LMVR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, MD 20852, USA
| | - Louis H Miller
- Laboratory of Malaria and Vector Research (LMVR), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, MD 20852, USA
| | - Takafumi Tsuboi
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Ehime 790-8577, Japan
| | - Eun-Taek Han
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do 200-701, Republic of Korea
| |
Collapse
|
17
|
Lee SK, Wang B, Han JH, Nyunt MH, Muh F, Chootong P, Ha KS, Park WS, Hong SH, Park JH, Han ET. Characterization of Pv92, a Novel Merozoite Surface Protein of Plasmodium vivax. THE KOREAN JOURNAL OF PARASITOLOGY 2016; 54:385-91. [PMID: 27658588 PMCID: PMC5040082 DOI: 10.3347/kjp.2016.54.4.385] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 05/29/2016] [Accepted: 05/29/2016] [Indexed: 11/27/2022]
Abstract
The discovery and understanding of antigenic proteins are essential for development of a vaccine against malaria. In Plasmodium falciparum, Pf92 have been characterized as a merozoite surface protein, and this protein is expressed at the late schizont stage, but no study of Pv92, the orthologue of Pf92 in P. vivax, has been reported. Thus, the protein structure of Pv92 was analyzed, and the gene sequence was aligned with that of other Plasmodium spp. using bioinformatics tools. The recombinant Pv92 protein was expressed and purified using bacterial expression system and used for immunization of mice to gain the polyclonal antibody and for evaluation of antigenicity by protein array. Also, the antibody against Pv92 was used for subcellular analysis by immunofluorescence assay. The Pv92 protein has a signal peptide and a sexual stage s48/45 domain, and the cysteine residues at the N-terminal of Pv92 were completely conserved. The N-terminal of Pv92 was successfully expressed as soluble form using a bacterial expression system. The antibody raised against Pv92 recognized the parasites and completely merged with PvMSP1-19, indicating that Pv92 was localized on the merozoite surface. Evaluation of the human humoral immune response to Pv92 indicated moderate antigenicity, with 65% sensitivity and 95% specificity by protein array. Taken together, the merozoite surface localization and antigenicity of Pv92 implicate that it might be involved in attachment and invasion of a merozoite to a new host cell or immune evasion during invasion process.
Collapse
Affiliation(s)
- Seong-Kyun Lee
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| | - Bo Wang
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, Korea.,Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Jin-Hee Han
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| | - Myat Htut Nyunt
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| | - Fauzi Muh
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| | - Patchanee Chootong
- Department of Clinical Microbiology and Applied Technology, Mahidol University, Bangkok, Thailand
| | - Kwon-Soo Ha
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| | - Won Sun Park
- Department of Physiology, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| | - Seok-Ho Hong
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| | - Jeong-Hyun Park
- Department of Anatomy, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| | - Eun-Taek Han
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, Korea
| |
Collapse
|
18
|
França CT, Hostetler JB, Sharma S, White MT, Lin E, Kiniboro B, Waltmann A, Darcy AW, Li Wai Suen CSN, Siba P, King CL, Rayner JC, Fairhurst RM, Mueller I. An Antibody Screen of a Plasmodium vivax Antigen Library Identifies Novel Merozoite Proteins Associated with Clinical Protection. PLoS Negl Trop Dis 2016; 10:e0004639. [PMID: 27182597 PMCID: PMC4868274 DOI: 10.1371/journal.pntd.0004639] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Accepted: 03/29/2016] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Elimination of Plasmodium vivax malaria would be greatly facilitated by the development of an effective vaccine. A comprehensive and systematic characterization of antibodies to P. vivax antigens in exposed populations is useful in guiding rational vaccine design. METHODOLOGY/PRINCIPAL FINDINGS In this study, we investigated antibodies to a large library of P. vivax entire ectodomain merozoite proteins in 2 Asia-Pacific populations, analysing the relationship of antibody levels with markers of current and cumulative malaria exposure, and socioeconomic and clinical indicators. 29 antigenic targets of natural immunity were identified. Of these, 12 highly-immunogenic proteins were strongly associated with age and thus cumulative lifetime exposure in Solomon Islanders (P<0.001-0.027). A subset of 6 proteins, selected on the basis of immunogenicity and expression levels, were used to examine antibody levels in plasma samples from a population of young Papua New Guinean children with well-characterized individual differences in exposure. This analysis identified a strong association between reduced risk of clinical disease and antibody levels to P12, P41, and a novel hypothetical protein that has not previously been studied, PVX_081550 (IRR 0.46-0.74; P<0.001-0.041). CONCLUSION/SIGNIFICANCE These data emphasize the benefits of an unbiased screening approach in identifying novel vaccine candidate antigens. Functional studies are now required to establish whether PVX_081550 is a key component of the naturally-acquired protective immune response, a biomarker of immune status, or both.
Collapse
Affiliation(s)
- Camila T. França
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Jessica B. Hostetler
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
- Malaria Programme, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Sumana Sharma
- Malaria Programme, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Michael T. White
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
- Center for Outbreak Analysis and Modelling, Department of Infectious Disease Epidemiology, Imperial College London, London, United Kingdom
| | - Enmoore Lin
- Vector Borne Diseases Unit, PNG Institute of Medical Research, Madang, Papua New Guinea
| | - Benson Kiniboro
- Vector Borne Diseases Unit, PNG Institute of Medical Research, Madang, Papua New Guinea
| | - Andreea Waltmann
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Andrew W. Darcy
- National Health Training & Research Institute, Ministry of Health, Honiara, Solomon Islands
| | - Connie S. N. Li Wai Suen
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Peter Siba
- Vector Borne Diseases Unit, PNG Institute of Medical Research, Madang, Papua New Guinea
| | - Christopher L. King
- Center for Global Health & Diseases, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Julian C. Rayner
- Malaria Programme, Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Rick M. Fairhurst
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Ivo Mueller
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
- ISGlobal, Barcelona Ctr. Int. Health Res. (CRESIB), Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
19
|
Chen JH, Chen SB, Wang Y, Ju C, Zhang T, Xu B, Shen HM, Mo XJ, Molina DM, Eng M, Liang X, Gardner MJ, Wang R, Hu W. An immunomics approach for the analysis of natural antibody responses to Plasmodium vivax infection. MOLECULAR BIOSYSTEMS 2016; 11:2354-63. [PMID: 26091354 DOI: 10.1039/c5mb00330j] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
High throughput immunomics is a powerful platform to discover potential targets of host immunity and develop diagnostic tests for infectious diseases. We screened the sera of Plasmodium vivax-exposed individuals to profile the antibody response to blood-stage antigens of P. vivax using a P. vivax protein microarray. A total of 1936 genes encoding the P. vivax proteins were expressed, printed and screened with sera from P. vivax-exposed individuals and normal subjects. Total of 151 (7.8% of the 1936 targets) highly immunoreactive antigens were identified, including five well-characterized antigens of P. vivax (ETRAMP11.2, Pv34, SUB1, RAP2 and MSP4). Among the highly immunoreactive antigens, 5 antigens were predicted as adhesins by MAAP, and 11 antigens were predicted as merozoite invasion-related proteins based on homology with P. falciparum proteins. There are 40 proteins that have serodiagnostic potential for antibody surveillance. These novel Plasmodium antigens identified provide the clues for understanding host immune response to P. vivax infection and the development of antibody surveillance tools.
Collapse
Affiliation(s)
- Jun-Hu Chen
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Key Laboratory of Parasite and Vector Biology of the Chinese Ministry of Health, WHO Collaborating Center for Malaria, Schistosomiasis and Filariasis, Shanghai 200025, People's Republic of China.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Hostetler JB, Sharma S, Bartholdson SJ, Wright GJ, Fairhurst RM, Rayner JC. A Library of Plasmodium vivax Recombinant Merozoite Proteins Reveals New Vaccine Candidates and Protein-Protein Interactions. PLoS Negl Trop Dis 2015; 9:e0004264. [PMID: 26701602 PMCID: PMC4689532 DOI: 10.1371/journal.pntd.0004264] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Accepted: 11/05/2015] [Indexed: 11/27/2022] Open
Abstract
Background A vaccine targeting Plasmodium vivax will be an essential component of any comprehensive malaria elimination program, but major gaps in our understanding of P. vivax biology, including the protein-protein interactions that mediate merozoite invasion of reticulocytes, hinder the search for candidate antigens. Only one ligand-receptor interaction has been identified, that between P. vivax Duffy Binding Protein (PvDBP) and the erythrocyte Duffy Antigen Receptor for Chemokines (DARC), and strain-specific immune responses to PvDBP make it a complex vaccine target. To broaden the repertoire of potential P. vivax merozoite-stage vaccine targets, we exploited a recent breakthrough in expressing full-length ectodomains of Plasmodium proteins in a functionally-active form in mammalian cells and initiated a large-scale study of P. vivax merozoite proteins that are potentially involved in reticulocyte binding and invasion. Methodology/Principal Findings We selected 39 P. vivax proteins that are predicted to localize to the merozoite surface or invasive secretory organelles, some of which show homology to P. falciparum vaccine candidates. Of these, we were able to express 37 full-length protein ectodomains in a mammalian expression system, which has been previously used to express P. falciparum invasion ligands such as PfRH5. To establish whether the expressed proteins were correctly folded, we assessed whether they were recognized by antibodies from Cambodian patients with acute vivax malaria. IgG from these samples showed at least a two-fold change in reactivity over naïve controls in 27 of 34 antigens tested, and the majority showed heat-labile IgG immunoreactivity, suggesting the presence of conformation-sensitive epitopes and native tertiary protein structures. Using a method specifically designed to detect low-affinity, extracellular protein-protein interactions, we confirmed a predicted interaction between P. vivax 6-cysteine proteins P12 and P41, further suggesting that the proteins are natively folded and functional. This screen also identified two novel protein-protein interactions, between P12 and PVX_110945, and between MSP3.10 and MSP7.1, the latter of which was confirmed by surface plasmon resonance. Conclusions/Significance We produced a new library of recombinant full-length P. vivax ectodomains, established that the majority of them contain tertiary structure, and used them to identify predicted and novel protein-protein interactions. As well as identifying new interactions for further biological studies, this library will be useful in identifying P. vivax proteins with vaccine potential, and studying P. vivax malaria pathogenesis and immunity. Trial Registration ClinicalTrials.gov NCT00663546 Plasmodium vivax causes malaria in millions of people each year, primarily in Southeast Asia and Central and South America. P. vivax has a dormant liver stage, which can lead to disease recurrence in infected individuals even in the absence of mosquito transmission. The development of vaccines that target blood-stage P. vivax parasites is therefore likely to be an essential component of any worldwide effort to eradicate malaria. Studying P. vivax is very difficult as this parasite grows poorly in the laboratory and invades only small numbers of young red blood cells in patients. Due to these and other challenges, only a handful of P. vivax proteins have been tested as potential vaccines. To generate more vaccine candidates, we expressed the entire ectodomains of 37 proteins that are predicted to be involved in P. vivax invasion of red blood cells. Antibodies from Cambodian patients with P. vivax malaria recognized heat-sensitive epitopes in the majority of these proteins, suggesting that they are natively folded. We also used the proteins to screen for both predicted and novel protein-protein interactions, confirming that the proteins are functional and further supporting their potential as vaccine candidates. As a new community resource, this P. vivax recombinant protein library will facilitate future studies of P. vivax pathogenesis and immunity, and greatly expands the list of candidate vaccine antigens.
Collapse
Affiliation(s)
- Jessica B. Hostetler
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
- Malaria Programme, Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Sumana Sharma
- Malaria Programme, Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
- Cell Surface Signalling Laboratory, Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - S. Josefin Bartholdson
- Malaria Programme, Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
- Cell Surface Signalling Laboratory, Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Gavin J. Wright
- Malaria Programme, Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
- Cell Surface Signalling Laboratory, Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Rick M. Fairhurst
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail: (RMF); (JCR)
| | - Julian C. Rayner
- Malaria Programme, Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
- * E-mail: (RMF); (JCR)
| |
Collapse
|
21
|
Cheng Y, Lu F, Lee SK, Kong DH, Ha KS, Wang B, Sattabongkot J, Tsuboi T, Han ET. Characterization of Plasmodium vivax Early Transcribed Membrane Protein 11.2 and Exported Protein 1. PLoS One 2015; 10:e0127500. [PMID: 26011536 PMCID: PMC4444142 DOI: 10.1371/journal.pone.0127500] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Accepted: 04/16/2015] [Indexed: 12/17/2022] Open
Abstract
In Plasmodium, the membrane of intracellular parasites is initially formed during invasion as an invagination of the red blood cell surface, which forms a barrier between the parasite and infected red blood cells in asexual blood stage parasites. The membrane proteins of intracellular parasites of Plasmodium species have been identified such as early-transcribed membrane proteins (ETRAMPs) and exported proteins (EXPs). However, there is little or no information regarding the intracellular parasite membrane in Plasmodium vivax. In the present study, recombinant PvETRAMP11.2 (PVX_003565) and PvEXP1 (PVX_091700) were expressed and evaluated antigenicity tests using sera from P. vivax-infected patients. A large proportion of infected individuals presented with IgG antibody responses against PvETRAMP11.2 (76.8%) and PvEXP1 (69.6%). Both of the recombinant proteins elicited high antibody titers capable of recognizing parasites of vivax malaria patients. PvETRAMP11.2 partially co-localized with PvEXP1 on the intracellular membranes of immature schizont. Moreover, they were also detected at the apical organelles of newly formed merozoites of mature schizont. We first proposed that these proteins might be synthesized in the preceding schizont stage, localized on the parasite membranes and apical organelles of infected erythrocytes, and induced high IgG antibody responses in patients.
Collapse
Affiliation(s)
- Yang Cheng
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, Maryland, United States of America
| | - Feng Lu
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
- Key Laboratory of Parasitic Disease Control and Prevention (Ministry of Health), Jiangsu Institute of Parasitic Diseases, Wuxi, Jiangsu Province, People’s Republic of China
- Jiangsu Provincial Key Laboratory of Parasite Molecular Biology, Jiangsu Institute of Parasitic Diseases, Wuxi, Jiangsu Province, People’s Republic of China
| | - Seong-Kyun Lee
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
| | - Deok-Hoon Kong
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
| | - Kwon-Soo Ha
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
| | - Bo Wang
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People’s Republic of China
| | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Takafumi Tsuboi
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Ehime, Japan
- * E-mail: (ETH); (TT)
| | - Eun-Taek Han
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
- * E-mail: (ETH); (TT)
| |
Collapse
|
22
|
Cheng Y, Li J, Ito D, Kong DH, Ha KS, Lu F, Wang B, Sattabongkot J, Lim CS, Tsuboi T, Han ET. Antigenicity and immunogenicity of PvRALP1, a novel Plasmodium vivax rhoptry neck protein. Malar J 2015; 14:186. [PMID: 25925592 PMCID: PMC4435652 DOI: 10.1186/s12936-015-0698-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 04/12/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Proteins secreted from the rhoptry in Plasmodium merozoites are associated with the formation of tight junctions and parasitophorous vacuoles during invasion of erythrocytes and are sorted within the rhoptry neck or bulb. Very little information has been obtained to date about Plasmodium vivax rhoptry-associated leucine (Leu) zipper-like protein 1 (PvRALP1; PVX_096245), a putative rhoptry protein. PvRALP1 contains a signal peptide, a glycine (Gly)/glutamate (Glu)-rich domain, and a Leu-rich domain, all of which are conserved in other Plasmodium species. METHODS Recombinant PvRALP1s were expressed as full-length protein without the signal peptide (PvRALP1-Ecto) and as truncated protein consisting of the Gly/Glu- and Leu-rich domains (PvRALP1-Tr) using the wheat germ cell-free expression system. The immunoreactivity to these two fragments of recombinant PvRALP1 protein in serum samples from P. vivax-infected patients and immunized mice, including analysis of immunoglobulin G (IgG) subclasses, was evaluated by enzyme-linked immunosorbent assay or protein microarray technology. The subcellular localization of PvRALP1 in blood stage parasites was also determined. RESULTS Recombinant PvRALP1-Ecto and PvRALP1-Tr proteins were successfully expressed, and in serum samples from P. vivax patients from the Republic of Korea, the observed immunoreactivities to these proteins had 58.9% and 55.4% sensitivity and 95.0% and 92.5% specificity, respectively. The response to PvRALP1 in humans was predominantly cytophilic antibodies (IgG1 and IgG3), but a balanced Th1/Th2 response was observed in mice. Unexpectedly, there was no significant inverse correlation between levels of parasitaemia and levels of antibody against either PvRALP1-Ecto (R2=0.11) or PvRALP1-Tr (R2=0.14) antigens. PvRALP1 was localized in the rhoptry neck of merozoites, and this was the first demonstration of the localization of this protein in P. vivax. CONCLUSIONS This study analysed the antigenicity and immunogenicity of PvRALP1 and suggested that PvRALP1 may be immunogenic in humans during parasite infection and might play an important role during invasion of P. vivax parasites.
Collapse
Affiliation(s)
- Yang Cheng
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Hyoja2-dong, Chuncheon, Gangwon-do, 200-701, Republic of Korea. .,Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, MD, 20852, USA.
| | - Jian Li
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Hyoja2-dong, Chuncheon, Gangwon-do, 200-701, Republic of Korea. .,Department of Parasitology, College of Basic Medicine, Hubei University of Medicine, Hubei, 442000, China.
| | - Daisuke Ito
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, MD, 20852, USA. .,Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Ehime, 790-8577, Japan.
| | - Deok-Hoon Kong
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, 200-701, Republic of Korea.
| | - Kwon-Soo Ha
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, 200-701, Republic of Korea.
| | - Feng Lu
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Hyoja2-dong, Chuncheon, Gangwon-do, 200-701, Republic of Korea. .,Key Laboratory of Parasitic Disease Control and Prevention (Ministry of Health), and Jiangsu Provincial Key Laboratory of Parasite Molecular Biology, Jiangsu Institute of Parasitic Diseases, Wuxi, Jiangsu Province, People's Republic of China.
| | - Bo Wang
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Hyoja2-dong, Chuncheon, Gangwon-do, 200-701, Republic of Korea. .,Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China.
| | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand.
| | - Chae Seung Lim
- Department of Laboratory Medicine, College of Medicine, Korea University Guro Hospital, Seoul, 152-703, Republic of Korea.
| | - Takafumi Tsuboi
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Ehime, 790-8577, Japan.
| | - Eun-Taek Han
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Hyoja2-dong, Chuncheon, Gangwon-do, 200-701, Republic of Korea.
| |
Collapse
|
23
|
Oguike MC, Sutherland CJ. Dimorphism in genes encoding sexual-stage proteins of Plasmodium ovale curtisi and Plasmodium ovale wallikeri. Int J Parasitol 2015; 45:449-54. [PMID: 25817462 PMCID: PMC4428623 DOI: 10.1016/j.ijpara.2015.02.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 02/09/2015] [Accepted: 02/09/2015] [Indexed: 01/19/2023]
Abstract
We present novel sequence data for putative sexual stage genes in Plasmodium ovale spp. Substantial dimorphism between the two Plasmodium ovale spp. is demonstrated by these genes. Our data provide further evidence for non-recombination between ovale spp. This information provides a platform for further research into the biology of ovale malaria.
Plasmodium ovale curtisi and Plasmodium ovale wallikeri are distinct species of malaria parasite which are sympatric throughout the tropics, except for the Americas. Despite this complete overlap in geographic range, these two species do not recombine. Although morphologically very similar, the two taxa must possess distinct characters which prevent recombination between them. We hypothesised that proteins required for sexual reproduction have sufficiently diverged between the two species to prevent recombination in any mosquito blood meal in which gametocytes of both species are ingested. In order to investigate possible barriers to inter-species mating between P. ovale curtisi and P. ovale wallikeri, homologues of genes encoding sexual stage proteins in other plasmodia were identified and compared between the two species. Database searches with motifs for 6-cysteine, Limulus Coagulation factor C domain-containing proteins and other relevant sexual stage proteins in the genus Plasmodium were performed in the available P. ovale curtisi partial genome database (Wellcome Trust Sanger Institute, UK). Sequence fragments obtained were used as the basis for PCR walking along each gene of interest in reference isolates of both P. ovale curtisi and P. ovale wallikeri. Sequence alignment of the homologues of each gene in each species showed complete dimorphism across all isolates. In conclusion, substantial divergence between sexual stage proteins in the two P. ovale spp. was observed, providing further evidence that these do not recombine in nature. Incompatibility of proteins involved in sexual development and fertilisation thus remains a plausible explanation for the observed lack of natural recombination between P. ovale curtisi and P. ovale wallikeri.
Collapse
Affiliation(s)
- Mary C Oguike
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, (LSHTM), UK.
| | - Colin J Sutherland
- Department of Immunology and Infection, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, (LSHTM), UK; Public Health England Malaria Reference Laboratory, Faculty of Infectious and Tropical Diseases, LSHTM, UK
| |
Collapse
|
24
|
Mo J, Li J. In silico analysis for structure, function and T-cell epitopes of a hypothetical conserved (HP-C) protein coded by PVX_092425 in Plasmodium vivax. Pathog Glob Health 2015; 109:61-7. [PMID: 25706099 DOI: 10.1179/2047773215y.0000000005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
OBJECTIVE Plasmodium spp. merozoite glycosylphosphatidylinositol-anchored proteins (GPI-APs) considered as protective immunogen in novel vaccines against malaria. To analyze the structure and function of a hypothetical conserved (HP-C) GPI-AP coded by gene PVX_092425 from Plasmodium vivax, and find its potential T-cell epitopes for further vivax malaria vaccine study. METHODS The structure, function and T-cell epitopes of the HP-C protein named Pvx_092425 were analyzed and predicted by online and offline bioinformatics software. RESULTS The bioinformatics data showed that the Pvx_092425 is an 830 amino acid (AA) long polypeptide encoded by five exons gene PVX_092425.It contains a pectin lyase-like superfamily, an AA repeats region, a cys-rich region and a transmembrane domain (TM) in C-terminal region. The alignment analysis drew it has a unique AA repeats region among Plasmodium spp. It was located in the cytoplasm, secretory system or cellular nucleus of P. vivax merozoite. For the sequence, the fragment of I823-V829 inserts in the interior side of the membrane, and M1--A812 belongs to the cytoplasmic tail. It has seven protein-protein binding sites. The peptides with the best predicted binding affinities were human leucocyte antigen (HLA) HLA-A*0203, HLA-DRB1*0101 and HLA- DRB1*0701.Among these predicted peptides, 582FLWDKALFD590 epitope interacted with HLA-DRB1*0101 allele showed best binding affinity compared to others. Structural analysis explained that the epitope fits well into the epitope-binding groove of HLA-DRB1*0101. CONCLUSIONS It proposes that the Pvx_092425 plays a key role during erythrocyte stage and generates information that is useful for development of blood-stage vaccine to block the merozoites invasion.
Collapse
|
25
|
Wang Y, Ma A, Chen SB, Yang YC, Chen JH, Yin MB. Genetic diversity and natural selection of three blood-stage 6-Cys proteins in Plasmodium vivax populations from the China-Myanmar endemic border. INFECTION GENETICS AND EVOLUTION 2014; 28:167-74. [PMID: 25266249 DOI: 10.1016/j.meegid.2014.09.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 09/12/2014] [Accepted: 09/21/2014] [Indexed: 11/30/2022]
Abstract
Pv12, Pv38 and Pv41, the three 6-Cys family proteins which are expressed in the blood-stage of vivax malaria, might be involved in merozoite invasion activity and thus be potential vaccine candidate antigens of Plasmodium vivax. However, little information is available concerning the genetic diversity and natural selection of these three proteins. In the present study, we analyzed the amino acid sequences of P. vivax blood-stage 6-Cys family proteins in comparison with the homologue proteins of Plasmodium cynomolgi strain B using bioinformatic methods. We also investigated genetic polymorphisms and natural selection of these three genes in P. vivax populations from the China-Myanmar endemic border. The three P. vivax blood-stage 6-Cys proteins were shown to possess a signal peptide at the N-terminus, containing two s48/45 domains, and Pv12 and Pv38 have a GPI-anchor motif at the C-terminus. Then, 22, 21 and 29 haplotypes of pv12, pv38 and pv41 were identified out of 45, 38 and 40 isolates, respectively. The dN/dS values for Domain II of pv38 and pv41 were 3.33880 and 5.99829, respectively, suggesting positive balancing selection for these regions. Meanwhile, the C-terminus of pv41 showed high nucleotide diversity, and Tajima's D test suggested that this fragment could be under positive balancing selection. Overall, our results have significant implications, providing a genetic basis for blood-stage malaria vaccine development based on these three 6-Cys proteins.
Collapse
Affiliation(s)
- Yue Wang
- Institute of Parasitic Diseases, Zhejiang Academy of Medical Sciences, Hangzhou 310013, Zhejiang, People's Republic of China
| | - An Ma
- Institute of Parasitic Diseases, Zhejiang Academy of Medical Sciences, Hangzhou 310013, Zhejiang, People's Republic of China
| | - Shen-Bo Chen
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Key Laboratory of Parasite and Vector Biology of the Chinese Ministry of Health, WHO Collaborating Center for Malaria, Schistosomiasis and Filariasis, Shanghai 200025, People's Republic of China
| | - Ying-Chao Yang
- Division of Parasitic Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control, Beijing 100050, People's Republic of China
| | - Jun-Hu Chen
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Key Laboratory of Parasite and Vector Biology of the Chinese Ministry of Health, WHO Collaborating Center for Malaria, Schistosomiasis and Filariasis, Shanghai 200025, People's Republic of China.
| | - Ming-Bo Yin
- Coastal Ecosystems Research Station of Yangtze River Estuary, Ministry of Education Key Laboratory for Biodiversity Science and Ecological Engineering, Institute of Biodiversity Science, Fudan University, Shanghai 200433, People's Republic of China.
| |
Collapse
|
26
|
Lu F, Li J, Wang B, Cheng Y, Kong DH, Cui L, Ha KS, Sattabongkot J, Tsuboi T, Han ET. Profiling the humoral immune responses to Plasmodium vivax infection and identification of candidate immunogenic rhoptry-associated membrane antigen (RAMA). J Proteomics 2014; 102:66-82. [PMID: 24607491 DOI: 10.1016/j.jprot.2014.02.029] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Revised: 02/05/2014] [Accepted: 02/23/2014] [Indexed: 11/17/2022]
Abstract
UNLABELLED Completion of sequencing of the Plasmodium vivax genome and transcriptome offers the chance to identify antigens among >5000 candidate proteins. To identify those P. vivax proteins that are immunogenic, a total of 152 candidate proteins (160 fragments) were expressed using a wheat germ cell-free system. The results of Western blot analysis showed that 92.5% (148/160) of the targets were expressed, and 96.6% (143/148) were in a soluble form with 67.7% of solubility rate. The proteins were screened by protein arrays with sera from 22 vivax malaria patients and 10 healthy individuals to confirm their immune profile, and 44 (27.5%, 44/160) highly reactive P. vivax antigens were identified. Overall, 5 candidates (rhoptry-associated membrane antigen [RAMA], Pv-fam-a and -b, EXP-1 and hypothetical protein PVX_084775) showed a positive reaction with >80% of patient sera, and 21 candidates with 50% to 80%. More than 23% of the highly immunoreactive proteins were hypothetical proteins, described for the first time in this study. One of the top immunogenic proteins, RAMA, was characterized and confirmed to be a serological marker of recent exposure to P. vivax infection. These novel immunoproteomes should greatly facilitate the identification of promising novel malaria antigens and may warrant further study. BIOLOGICAL SIGNIFICANCE The establishment of high-throughput cloning and expression systems has permitted the construction of protein arrays for proteome-wide study of Plasmodium vivax. In this study, high-throughput screening assays have been applied to investigate blood stage-specific immune proteomes from P. vivax. We identified 44 antigenic proteins from the 152 putative candidates, more than 23% of which were hypothetical proteins described for the first time in this study. In addition, PvRAMA was characterized further and confirmed to be a serological marker of exposure to infections. The expression of one-third of the selected antigenic genes were shifted between P. vivax and Plasmodium falciparum, suggesting that these genes may represent important factors associated with P. vivax selectivity for young erythrocytes and/or with immune evasion. These novel immune proteomes of the P. vivax blood stage provide a baseline for further prospective serological marker studies in malaria. These methods could be used to determine immunodominant candidate antigens from the P. vivax genome.
Collapse
Affiliation(s)
- Feng Lu
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chunchon, Gangwon-do, Republic of Korea; Jiangsu Institute of Parasitic Diseases, Key Laboratory on Technology for Parasitic Disease Prevention and Control, Ministry of Health, Wuxi, Jiangsu, People's Republic of China
| | - Jian Li
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chunchon, Gangwon-do, Republic of Korea; Jiangsu Institute of Parasitic Diseases, Key Laboratory on Technology for Parasitic Disease Prevention and Control, Ministry of Health, Wuxi, Jiangsu, People's Republic of China
| | - Bo Wang
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chunchon, Gangwon-do, Republic of Korea
| | - Yang Cheng
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chunchon, Gangwon-do, Republic of Korea
| | - Deok-Hoon Kong
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chunchon, Gangwon-do, Republic of Korea
| | - Liwang Cui
- Department of Entomology, The Pennsylvania State University, University Park, PA, USA
| | - Kwon-Soo Ha
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chunchon, Gangwon-do, Republic of Korea
| | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Takafumi Tsuboi
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Ehime, Japan.
| | - Eun-Taek Han
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chunchon, Gangwon-do, Republic of Korea.
| |
Collapse
|
27
|
Arumugam TU, Ito D, Takashima E, Tachibana M, Ishino T, Torii M, Tsuboi T. Application of wheat germ cell-free protein expression system for novel malaria vaccine candidate discovery. Expert Rev Vaccines 2013; 13:75-85. [DOI: 10.1586/14760584.2014.861747] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
28
|
Cheng Y, Ito D, Sattabongkot J, Lim CS, Kong DH, Ha KS, Wang B, Tsuboi T, Han ET. Serological responses to a soluble recombinant chimeric Plasmodium vivax circumsporozoite protein in VK210 and VK247 population. Malar J 2013; 12:323. [PMID: 24034268 PMCID: PMC3847697 DOI: 10.1186/1475-2875-12-323] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 09/11/2013] [Indexed: 12/01/2022] Open
Abstract
Background Circumsporozoite protein (CSP) is essential for sporozoite formation and sporozoite invasion into human hepatocyte. Previously, a recombinant P. vivax CSP based on chimeric repeats (rPvCSP-c) representing two major alleles VK210 and VK247 within central region has been designed. Naturally acquired humoral immune responses study show that antigenicity of rPvCSP-c was much higher than that of native strain. However, the serologic reactivity of rPvCSP-c was still unclear in detail. Methods In present study, recognition of rPvCSP-c in vivax malaria typed VK210 and VK247 alleles was assessed. VK210 typed and VK247 typed sera from adult residents reacted specifically with rPvCSP-c using protein array and immunoblot assay. Additionally, anti-rPvCSP-c serum recognized the fixed VK210 and VK247 sporozoites by immunofluorescence assay. Furthermore, statistic analysis was performed for correlational detection. Results The rPvCSP-c reacted with both VK210 typed and VK247 typed P. vivax infected patient sera and anti-rPvCSP-c immune serum also reacted with VK210 and VK247 sporozoite parasites of P. vivax specifically. There was a positive correlation between increased antibody level, age of patients and also associated with pvcsp repeat number, although the level of responses did vary considerably in their reactivity to the rPvCSP-c from negative to very high level within each age group. Conclusions These data confirmed the serologic reactivity of the novel rPvCSP-c in exposed both VK210 and VK247 populations. These results strongly suggested that this recombinant CSP was biologically active and potently immunogenic across major strains and raised the prospect that this protein could be used as serologic marker.
Collapse
Affiliation(s)
- Yang Cheng
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon, Gangwon-do 200-701, Republic of Korea.
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Cheng Y, Lu F, Tsuboi T, Han ET. Characterization of a novel merozoite surface protein of Plasmodium vivax, Pv41. Acta Trop 2013; 126:222-8. [PMID: 23499861 DOI: 10.1016/j.actatropica.2013.03.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Revised: 02/28/2013] [Accepted: 03/01/2013] [Indexed: 10/27/2022]
Abstract
Since the genome of Plasmodium vivax was sequenced, few proteins have been characterized as highly immunogenic and candidates for inclusion in a vivax malaria vaccine. The P. vivax 41 (Pv41) protein has a signal peptide, one glutamate-rich domain in its central region, and two sexual stage s48/45 domains, and is characterized as a gametocyte surface protein; however, this protein may be expressed principally on the merozoite surface of parasites. The previous study reported the transcription, blood-stage expression, and subcellular localization of Pv41 within the parasite. In this study, the recombinant Pv41 protein was expressed as a soluble form, of a molecular mass ~44 kDa, by a cell-free expression system and was specifically recognized by animal immune sera and vivax patient sera. Evaluation of the human humoral immune response to Pv41 indicated a high immunogenicity, with 62.5% sensitivity and 95% specificity, by protein array. Immunofluorescence assays (IFA) using polyclonal anti-Pv41 antibodies showed that Pv41 was localized on the merozoite surface. The high immunogenicity of Pv41 indicates its potential as a vivax malaria vaccine candidate antigen, particularly in light of its location on the merozoite surface of the parasite.
Collapse
|
30
|
Tonkin ML, Arredondo SA, Loveless BC, Serpa JJ, Makepeace KA, Sundar N, Petrotchenko EV, Miller LH, Grigg ME, Boulanger MJ. Structural and Biochemical Characterization of Plasmodium falciparum 12 (Pf12) Reveals a Unique Interdomain Organization and the Potential for an Antiparallel Arrangement with Pf41. J Biol Chem 2013; 288:12805-17. [DOI: 10.1074/jbc.m113.455667] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
31
|
The Plasmodium vivax merozoite surface protein 1 paralog is a novel erythrocyte-binding ligand of P. vivax. Infect Immun 2013; 81:1585-95. [PMID: 23460511 DOI: 10.1128/iai.01117-12] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Merozoite surface protein 1 of Plasmodium vivax (PvMSP1), a glycosylphosphatidylinositol-anchored protein (GPI-AP), is a malaria vaccine candidate for P. vivax. The paralog of PvMSP1, named P. vivax merozoite surface protein 1 paralog (PvMSP1P; PlasmoDB PVX_099975), was recently identified and predicted as a GPI-AP. The similarities in genetic structural characteristics between PvMSP1 and PvMSP1P (e.g., size of open reading frames, two epidermal growth factor-like domains, and GPI anchor motif in the C terminus) led us to study this protein. In the present study, different regions of the PvMSP1P protein, demarcated based on the processed forms of PvMSP1, were expressed successfully as recombinant proteins [i.e., 83 (A, B, and C), 30, 38, 42, 33, and 19 fragments]. We studied the naturally acquired immune response against each fragment of recombinant PvMSP1P and the potential ability of each fragment to bind erythrocytes. The N-terminal fragment (83A) and two C-terminal fragments (33 and 19) reacted strongly with sera from P. vivax-infected patients, with 50 to 68% sensitivity and 95 to 96% specificity, respectively. Due to colocalization of PvMSP1P with PvMSP1, we supposed that PvMSP1P plays a similar role as PvMSP1 during erythrocyte invasion. An in vitro cytoadherence assay showed that PvMSP1P, especially the 19-kDa C-terminal region, could bind to erythrocytes. We also found that human sera from populations naturally exposed to vivax malaria and antisera obtained by immunization using the recombinant molecule PvMSP1P-19 inhibited in vitro binding of human erythrocytes to PvMSP1P-19. These results provide further evidence that the PvMSP1P might be an essential parasite adhesion molecule in the P. vivax merozoite and is a potential vaccine candidate against P. vivax.
Collapse
|
32
|
Wang B, Lu F, Cheng Y, Li J, Ito D, Sattabongkot J, Tsuboi T, Han ET. Identification and characterization of the Plasmodium falciparum RhopH2 ortholog in Plasmodium vivax. Parasitol Res 2012; 112:585-93. [PMID: 23097184 DOI: 10.1007/s00436-012-3170-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2012] [Accepted: 10/08/2012] [Indexed: 11/28/2022]
Abstract
Plasmodium vivax is one of the most important human malaria species that is geographically widely endemic and potentially affects a larger number of people than its more notorious cousin, Plasmodium falciparum. During invasion of red blood cells, the parasite requires the intervention of high molecular weight complex rhoptry proteins (RhopH) that are also essential for cytoadherence. PfRhopH2, a member of the RhopH multigene family, has been characterized as being crucial during P. falciparum infection. This study describes identifying and characterizing the pfrhoph2 orthologous gene in P. vivax (hereinafter named pvrhoph2). The PvRhopH2 is a 1,369-amino acid polypeptide encoded by PVX_099930 gene, for which orthologous genes have been identified in other Plasmodium species by bioinformatic approaches. Both P. falciparum and P. vivax genes contain nine introns, and there is a high degree of similarity between the deduced amino acid sequences of the two proteins. Moreover, PvRhopH2 contains a signal peptide at its N-terminus and 12 cysteines predominantly in its C-terminal half. PvRhopH2 is localized in one of the apical organelles of the merozoite, the rhoptry, and the localization pattern is similar to that of PfRhopH2 in P. falciparum. The recombinant PvRhopH2 protein is recognized by serum antibodies of patients naturally exposed to P. vivax, suggesting that PvRhopH2 is immunogenic in humans.
Collapse
Affiliation(s)
- Bo Wang
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Hyoja2-dong, Chuncheon, Gangwon-do 200-701, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Li J, Han ET. Dissection of the Plasmodium vivax reticulocyte binding-like proteins (PvRBPs). Biochem Biophys Res Commun 2012; 426:1-6. [DOI: 10.1016/j.bbrc.2012.08.055] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Accepted: 08/11/2012] [Indexed: 01/08/2023]
|
34
|
Goldston AM, Powell RR, Temesvari LA. Sink or swim: lipid rafts in parasite pathogenesis. Trends Parasitol 2012; 28:417-26. [PMID: 22906512 DOI: 10.1016/j.pt.2012.07.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Revised: 07/16/2012] [Accepted: 07/17/2012] [Indexed: 12/26/2022]
Abstract
Lipid rafts, sterol- and sphingolipid-rich membrane microdomains, have been extensively studied in mammalian cells. Recently, lipid rafts have been shown to control virulence in a variety of parasites including Entamoeba histolytica, Giardia intestinalis, Leishmania spp., Plasmodium spp., Toxoplasma gondii, and Trypanosoma spp. Parasite rafts regulate adhesion to host and invasion, and parasite adhesion molecules often localize to rafts. Parasite rafts also control vesicle trafficking, motility, and cell signaling. Parasites disrupt host cell rafts; the dysregulation of host membrane function facilitates the establishment of infection and evasion of the host immune system. Discerning the mechanism by which lipid rafts regulate parasite pathogenesis is essential to our understanding of virulence. Such insight may guide the development of new drugs for disease management.
Collapse
Affiliation(s)
- Amanda M Goldston
- Department of Genetics and Biochemistry, Clemson University, Clemson, SC 29634, USA
| | | | | |
Collapse
|
35
|
The GPI-anchored 6-Cys protein Pv12 is present in detergent-resistant microdomains of Plasmodium vivax blood stage schizonts. Protist 2012; 164:37-48. [PMID: 22554829 DOI: 10.1016/j.protis.2012.03.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2011] [Revised: 03/16/2012] [Accepted: 03/31/2012] [Indexed: 11/21/2022]
Abstract
Plasmodium vivax malaria remains one of the tropical diseases causing an enormous burden on global public health. Several proteins located on this parasite species' merozoite surface have been considered the most suitable antigens for being included in an anti-malarial vaccine, given the functional role they play during the parasite's interaction with red blood cells. The present study identifies and characterizes the P. vivax Pv12 surface protein which was evaluated by using molecular biology and immunochemistry assays; its antigenic potential was also examined in natural and experimental P. vivax malaria infections. The P. vivax VCG-1 strain Pv12 gene encodes a 362 amino acid-long protein exhibiting a signal peptide, a glycosylphosphatidylinositol (GPI) anchor sequence and two 6-Cys domains. The presence of the Pv12 protein on the parasite's surface and its association with detergent-resistant membrane complexes, together with its antigenic potential, supports the notion that this antigen could play an important role as a red blood cell binding ligand. Further studies aimed at establishing the immunogenicity and protection-inducing ability of the Pv12 protein or its products in the Aotus experimental model are thus suggested.
Collapse
|