1
|
Lokole PB, Byamungu GG, Mutwale PK, Ngombe NK, Mudogo CN, Krause RWM, Nkanga CI. Plant-based nanoparticles targeting malaria management. Front Pharmacol 2024; 15:1440116. [PMID: 39185312 PMCID: PMC11341498 DOI: 10.3389/fphar.2024.1440116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 07/18/2024] [Indexed: 08/27/2024] Open
Abstract
Malaria is one of the most devastating diseases across the globe, particularly in low-income countries in Sub-Saharan Africa. The increasing incidence of malaria morbidity is mainly due to the shortcomings of preventative measures such as the lack of vaccines and inappropriate control over the parasite vector. Additionally, high mortality rates arise from therapeutic failures due to poor patient adherence and drug resistance development. Although the causative pathogen (Plasmodium spp.) is an intracellular parasite, the recommended antimalarial drugs show large volumes of distribution and low-to no-specificity towards the host cell. This leads to severe side effects that hamper patient compliance and promote the emergence of drug-resistant strains. Recent research efforts are promising to enable the discovery of new antimalarial agents; however, the lack of efficient means to achieve targeted delivery remains a concern, given the risk of further resistance development. New strategies based on green nanotechnologies are a promising avenue for malaria management due to their potential to eliminate malaria vectors (Anopheles sp.) and to encapsulate existing and emerging antimalarial agents and deliver them to different target sites. In this review we summarized studies on the use of plant-derived nanoparticles as cost-effective preventative measures against malaria parasites, starting from the vector stage. We also reviewed plant-based nanoengineering strategies to target malaria parasites, and further discussed the site-specific delivery of natural products using ligand-decorated nanoparticles that act through receptors on the host cells or malaria parasites. The exploration of traditionally established plant medicines, surface-engineered nanoparticles and the molecular targets of parasite/host cells may provide valuable insights for future discovery of antimalarial drugs and open new avenues for advancing science toward the goal of malaria eradication.
Collapse
Affiliation(s)
- Pathy B. Lokole
- Centre de Recherche en Nanotechnologies Appliquées aux Produits Naturels (CReNAPN), Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmaceutical Sciences, University of Kinshasa, Kinshasa, Democratic Republic of the Congo
- Centre d’Etudes des Substances Naturelles d’Origine Végétale (CESNOV), Faculty of Pharmaceutical Sciences, University of Kinshasa, Kinshasa, Democratic Republic of the Congo
- Center for Chemico- and Bio-Medicinal Research (CCBR), Department of Chemistry, Faculty of Sciences, Rhodes University, Grahamstown, Eastern Cape, South Africa
| | - Galilée G. Byamungu
- Centre de Recherche en Nanotechnologies Appliquées aux Produits Naturels (CReNAPN), Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmaceutical Sciences, University of Kinshasa, Kinshasa, Democratic Republic of the Congo
- Center for Chemico- and Bio-Medicinal Research (CCBR), Department of Chemistry, Faculty of Sciences, Rhodes University, Grahamstown, Eastern Cape, South Africa
- Department of Chemistry, Faculty of Sciences and Technology, University of Kinshasa, Kinshasa, Democratic Republic of the Congo
| | - Paulin K. Mutwale
- Centre de Recherche en Nanotechnologies Appliquées aux Produits Naturels (CReNAPN), Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmaceutical Sciences, University of Kinshasa, Kinshasa, Democratic Republic of the Congo
- Centre d’Etudes des Substances Naturelles d’Origine Végétale (CESNOV), Faculty of Pharmaceutical Sciences, University of Kinshasa, Kinshasa, Democratic Republic of the Congo
| | - Nadège K. Ngombe
- Centre de Recherche en Nanotechnologies Appliquées aux Produits Naturels (CReNAPN), Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmaceutical Sciences, University of Kinshasa, Kinshasa, Democratic Republic of the Congo
- Centre d’Etudes des Substances Naturelles d’Origine Végétale (CESNOV), Faculty of Pharmaceutical Sciences, University of Kinshasa, Kinshasa, Democratic Republic of the Congo
| | - Celestin N. Mudogo
- Unit of Molecular Biology, Department of Basic Sciences, Faculty of Medicine, University of Kinshasa, Kinshasa, Democratic Republic of the Congo
| | - Rui W. M. Krause
- Center for Chemico- and Bio-Medicinal Research (CCBR), Department of Chemistry, Faculty of Sciences, Rhodes University, Grahamstown, Eastern Cape, South Africa
| | - Christian I. Nkanga
- Centre de Recherche en Nanotechnologies Appliquées aux Produits Naturels (CReNAPN), Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmaceutical Sciences, University of Kinshasa, Kinshasa, Democratic Republic of the Congo
| |
Collapse
|
2
|
Cao Y, Hayashi CTH, Kumar N. A Novel Ex Vivo Assay to Evaluate Functional Effectiveness of Plasmodium vivax Transmission-Blocking Vaccine Using Pvs25 Transgenic Plasmodium berghei. J Infect Dis 2024; 229:1894-1903. [PMID: 38408353 PMCID: PMC11175679 DOI: 10.1093/infdis/jiae102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 01/20/2024] [Accepted: 02/22/2024] [Indexed: 02/28/2024] Open
Abstract
BACKGROUND Plasmodium falciparum and Plasmodium vivax account for >90% global malaria burden. Transmission intervention strategies encompassing transmission-blocking vaccines (TBV) and drugs represent ideal public health tools to eliminate malaria at the population level. The availability of mature P. falciparum gametocytes through in vitro culture has facilitated development of a standard membrane feeding assay to assess efficacy of transmission interventions against P. falciparum. The lack of in vitro culture for P. vivax has significantly hampered similar progress on P. vivax and limited studies have been possible using blood from infected patients in endemic areas. The ethical and logistical limitations of on-time access to blood from patients have impeded the development of P. vivax TBVs. METHODS Transgenic murine malaria parasites (Plasmodium berghei) expressing TBV candidates offer a promising alternative for evaluation of P. vivax TBVs through in vivo studies in mice, and ex vivo membrane feeding assay (MFA). RESULTS We describe the development of transmission-competent transgenic TgPbvs25 parasites and optimization of parameters to establish an ex vivo MFA to evaluate P. vivax TBV based on Pvs25 antigen. CONCLUSIONS The MFA is expected to expedite Pvs25-based TBV development without dependence on blood from P. vivax-infected patients in endemic areas for evaluation.
Collapse
Affiliation(s)
- Yi Cao
- Department of Global Health, Milken Institute School of Public Health, George Washington University, Washington, District of Columbia, USA
| | - Clifford T H Hayashi
- Department of Global Health, Milken Institute School of Public Health, George Washington University, Washington, District of Columbia, USA
| | - Nirbhay Kumar
- Department of Global Health, Milken Institute School of Public Health, George Washington University, Washington, District of Columbia, USA
| |
Collapse
|
3
|
Kong WZ, Zhang HY, Sun YF, Song J, Jiang J, Cui HY, Zhang Y, Han S, Cheng Y. Plasmodium vivax tryptophan-rich antigen reduces type I collagen secretion via the NF-κBp65 pathway in splenic fibroblasts. Parasit Vectors 2024; 17:239. [PMID: 38802961 PMCID: PMC11131192 DOI: 10.1186/s13071-024-06264-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 03/26/2024] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND The spleen plays a critical role in the immune response against malaria parasite infection, where splenic fibroblasts (SFs) are abundantly present and contribute to immune function by secreting type I collagen (collagen I). The protein family is characterized by Plasmodium vivax tryptophan-rich antigens (PvTRAgs), comprising 40 members. PvTRAg23 has been reported to bind to human SFs (HSFs) and affect collagen I levels. Given the role of type I collagen in splenic immune function, it is important to investigate the functions of the other members within the PvTRAg protein family. METHODS Protein structural prediction was conducted utilizing bioinformatics analysis tools and software. A total of 23 PvTRAgs were successfully expressed and purified using an Escherichia coli prokaryotic expression system, and the purified proteins were used for co-culture with HSFs. The collagen I levels and collagen-related signaling pathway protein levels were detected by immunoblotting, and the relative expression levels of inflammatory factors were determined by quantitative real-time PCR. RESULTS In silico analysis showed that P. vivax has 40 genes encoding the TRAg family. The C-terminal region of all PvTRAgs is characterized by the presence of a domain rich in tryptophan residues. A total of 23 recombinant PvTRAgs were successfully expressed and purified. Only five PvTRAgs (PvTRAg5, PvTRAg16, PvTRAg23, PvTRAg30, and PvTRAg32) mediated the activation of the NF-κBp65 signaling pathway, which resulted in the production of inflammatory molecules and ultimately a significant reduction in collagen I levels in HSFs. CONCLUSIONS Our research contributes to the expansion of knowledge regarding the functional role of PvTRAgs, while it also enhances our understanding of the immune evasion mechanisms utilized by parasites.
Collapse
Affiliation(s)
- Wei-Zhong Kong
- Laboratory of Pathogen Infection and Immunity, Department of Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China
| | - Hang-Ye Zhang
- Laboratory of Pathogen Infection and Immunity, Department of Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China
- Case Room, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| | - Yi-Fan Sun
- Laboratory of Pathogen Infection and Immunity, Department of Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China
- Department of Laboratory Medicine, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, People's Republic of China
| | - Jing Song
- Department of Obstetrics and Gynecology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Jian Jiang
- Wuxi Red Cross Blood Center, Wuxi, 214000, China
| | - Heng-Yuan Cui
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China
| | - Yu Zhang
- Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China
| | - Su Han
- Laboratory of Pathogen Infection and Immunity, Department of Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China.
| | - Yang Cheng
- Laboratory of Pathogen Infection and Immunity, Department of Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China.
| |
Collapse
|
4
|
Tottey S, Shoji Y, Mark Jones R, Musiychuk K, Chichester JA, Miura K, Zhou L, Lee SM, Plieskatt J, Wu Y, Long CA, Streatfield SJ, Yusibov V. Engineering of a plant-produced virus-like particle to improve the display of the Plasmodium falciparum Pfs25 antigen and transmission-blocking activity of the vaccine candidate. Vaccine 2023; 41:938-944. [PMID: 36585278 PMCID: PMC9888754 DOI: 10.1016/j.vaccine.2022.12.048] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 12/04/2022] [Accepted: 12/19/2022] [Indexed: 12/29/2022]
Abstract
Malaria kills around 409,000 people a year, mostly children under the age of five. Malaria transmission-blocking vaccines work to reduce malaria prevalence in a community and have the potential to be part of a multifaceted approach required to eliminate the parasites causing the disease. Pfs25 is a leading malaria transmission-blocking antigen and has been successfully produced in a plant expression system as both a subunit vaccine and as a virus-like particle. This study demonstrates an improved version of the virus-like particle antigen display molecule by eliminating known protease sites from the prior A85 variant. This re-engineered molecule, termed B29, displays three times the number of Pfs25 antigens per virus-like particle compared to the original Pfs25 virus-like particle. An improved purification scheme was also developed, resulting in a substantially higher yield and improved purity. The molecule was evaluated in a mouse model and found to induce improved transmission-blocking activity at lower doses and longer durations than the original molecule.
Collapse
Affiliation(s)
- Stephen Tottey
- Fraunhofer USA Center Mid-Atlantic, Biotechnology Division, 9 Innovation Way, Newark, DE 19711, USA
| | - Yoko Shoji
- Fraunhofer USA Center Mid-Atlantic, Biotechnology Division, 9 Innovation Way, Newark, DE 19711, USA
| | - R Mark Jones
- Fraunhofer USA Center Mid-Atlantic, Biotechnology Division, 9 Innovation Way, Newark, DE 19711, USA
| | - Konstantin Musiychuk
- Fraunhofer USA Center Mid-Atlantic, Biotechnology Division, 9 Innovation Way, Newark, DE 19711, USA
| | - Jessica A Chichester
- Fraunhofer USA Center Mid-Atlantic, Biotechnology Division, 9 Innovation Way, Newark, DE 19711, USA
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway, Rockville, MD 20852, USA
| | - Luwen Zhou
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway, Rockville, MD 20852, USA
| | - Shwu-Maan Lee
- PATH's Malaria Vaccine Initiative, Washington, DC 20001, USA
| | | | - Yimin Wu
- PATH's Malaria Vaccine Initiative, Washington, DC 20001, USA
| | - Carole A Long
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway, Rockville, MD 20852, USA
| | - Stephen J Streatfield
- Fraunhofer USA Center Mid-Atlantic, Biotechnology Division, 9 Innovation Way, Newark, DE 19711, USA.
| | - Vidadi Yusibov
- Fraunhofer USA Center Mid-Atlantic, Biotechnology Division, 9 Innovation Way, Newark, DE 19711, USA
| |
Collapse
|
5
|
da Veiga GTS, Moriggi MR, Vettorazzi JF, Müller-Santos M, Albrecht L. Plasmodium vivax vaccine: What is the best way to go? Front Immunol 2023; 13:910236. [PMID: 36726991 PMCID: PMC9885200 DOI: 10.3389/fimmu.2022.910236] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 12/23/2022] [Indexed: 01/18/2023] Open
Abstract
Malaria is one of the most devastating human infectious diseases caused by Plasmodium spp. parasites. A search for an effective and safe vaccine is the main challenge for its eradication. Plasmodium vivax is the second most prevalent Plasmodium species and the most geographically distributed parasite and has been neglected for decades. This has a massive gap in knowledge and consequently in the development of vaccines. The most significant difficulties in obtaining a vaccine against P. vivax are the high genetic diversity and the extremely complex life cycle. Due to its complexity, studies have evaluated P. vivax antigens from different stages as potential targets for an effective vaccine. Therefore, the main vaccine candidates are grouped into preerythrocytic stage vaccines, blood-stage vaccines, and transmission-blocking vaccines. This review aims to support future investigations by presenting the main findings of vivax malaria vaccines to date. There are only a few P. vivax vaccines in clinical trials, and thus far, the best protective efficacy was a vaccine formulated with synthetic peptide from a circumsporozoite protein and Montanide ISA-51 as an adjuvant with 54.5% efficacy in a phase IIa study. In addition, the majority of P. vivax antigen candidates are polymorphic, induce strain-specific and heterogeneous immunity and provide only partial protection. Nevertheless, immunization with recombinant proteins and multiantigen vaccines have shown promising results and have emerged as excellent strategies. However, more studies are necessary to assess the ideal vaccine combination and test it in clinical trials. Developing a safe and effective vaccine against vivax malaria is essential for controlling and eliminating the disease. Therefore, it is necessary to determine what is already known to propose and identify new candidates.
Collapse
Affiliation(s)
- Gisele Tatiane Soares da Veiga
- Laboratory of Apicomplexan Parasites Research, Carlos Chagas Institute, Oswaldo Cruz Foundation (FIOCRUZ), Curitiba, Brazil,Nitrogen Fixation Laboratory, Department of Biochemistry and Molecular Biology, Federal University of Paraná (UFPR), Curitiba, Brazil
| | | | | | - Marcelo Müller-Santos
- Nitrogen Fixation Laboratory, Department of Biochemistry and Molecular Biology, Federal University of Paraná (UFPR), Curitiba, Brazil
| | - Letusa Albrecht
- Laboratory of Apicomplexan Parasites Research, Carlos Chagas Institute, Oswaldo Cruz Foundation (FIOCRUZ), Curitiba, Brazil,*Correspondence: Letusa Albrecht,
| |
Collapse
|
6
|
Kar S, Sinha A. Plasmodium vivax Duffy Binding Protein-Based Vaccine: a Distant Dream. Front Cell Infect Microbiol 2022; 12:916702. [PMID: 35909975 PMCID: PMC9325973 DOI: 10.3389/fcimb.2022.916702] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 06/21/2022] [Indexed: 11/13/2022] Open
Abstract
The neglected but highly prevalent Plasmodium vivax in South-east Asia and South America poses a great challenge, with regards to long-term in-vitro culturing and heavily limited functional assays. Such visible challenges as well as narrowed progress in development of experimental research tools hinders development of new drugs and vaccines. The leading vaccine candidate antigen Plasmodium vivax Duffy Binding Protein (PvDBP), is essential for reticulocyte invasion by binding to its cognate receptor, the Duffy Antigen Receptor for Chemokines (DARC), on the host’s reticulocyte surface. Despite its highly polymorphic nature, the amino-terminal cysteine-rich region II of PvDBP (PvDBPII) has been considered as an attractive target for vaccine-mediated immunity and has successfully completed the clinical trial Phase 1. Although this molecule is an attractive vaccine candidate against vivax malaria, there is still a question on its viability due to recent findings, suggesting that there are still some aspects which needs to be looked into further. The highly polymorphic nature of PvDBPII and strain-specific immunity due to PvDBPII allelic variation in Bc epitopes may complicate vaccine efficacy. Emergence of various blood-stage antigens, such as PvRBP, PvEBP and supposedly many more might stand in the way of attaining full protection from PvDBPII. As a result, there is an urgent need to assess and re-assess various caveats connected to PvDBP, which might help in designing a long-term promising vaccine for P. vivax malaria. This review mainly deals with a bunch of rising concerns for validation of DBPII as a vaccine candidate antigen for P. vivax malaria.
Collapse
|
7
|
Wang PP, Jiang X, Zhu L, Zhou D, Hong M, He L, Chen L, Yao S, Zhao Y, Chen G, Wang C, Cui L, Cao Y, Zhu X. A G-Protein-Coupled Receptor Modulates Gametogenesis via PKG-Mediated Signaling Cascade in Plasmodium berghei. Microbiol Spectr 2022; 10:e0015022. [PMID: 35404079 PMCID: PMC9045217 DOI: 10.1128/spectrum.00150-22] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 03/20/2022] [Indexed: 12/13/2022] Open
Abstract
Gametogenesis is essential for malaria parasite transmission, but the molecular mechanism of this process remains to be refined. Here, we identified a G-protein-coupled receptor 180 (GPR180) that plays a critical role in signal transduction during gametogenesis in Plasmodium. The P. berghei GPR180 was predominantly expressed in gametocytes and ookinetes and associated with the plasma membrane in female gametes and ookinetes. Knockout of pbgpr180 (Δpbgpr180) had no noticeable effect on blood-stage development but impaired gamete formation and reduced transmission of the parasites to mosquitoes. Transcriptome analysis revealed that a large proportion of the dysregulated genes in the Δpbgpr180 gametocytes had assigned functions in cyclic nucleotide signal transduction. In the Δpbgpr180 gametocytes, the intracellular cGMP level was significantly reduced, and the cytosolic Ca2+ mobilization showed a delay and a reduction in the magnitude during gametocyte activation. These results suggest that PbGPR180 functions upstream of the cGMP-protein kinase G-Ca2+ signaling pathway. In line with this functional prediction, the PbGPR180 protein was found to interact with several transmembrane transporter proteins and the small GTPase Rab6 in activated gametocytes. Allele replacement of pbgpr180 with the P. vivax ortholog pvgpr180 showed equal competence of the transgenic parasite in sexual development, suggesting functional conservation of this gene in Plasmodium spp. Furthermore, an anti-PbGPR180 monoclonal antibody and the anti-PvGPR180 serum possessed robust transmission-blocking activities. These results indicate that GPR180 is involved in signal transduction during gametogenesis in malaria parasites and is a promising target for blocking parasite transmission. IMPORTANCE Environmental changes from humans to mosquitoes activate gametogenesis of the malaria parasite, an obligative process for parasite transmission, but how the signals are relayed remains poorly understood. Here, we show the identification of a Plasmodium G-protein-coupled receptor, GPR180, and the characterization of its function in gametogenesis. In P. berghei, GPR180 is dispensable for asexual development and gametocytogenesis, but its deletion impairs gametogenesis and reduces transmission to mosquitoes. GPR180 appears to function upstream of the cGMP-protein kinase G-Ca2+ signaling pathway and is required for the maximum activity of this pathway. Genetic complementation shows that the GPR180 ortholog from the human malaria parasite P. vivax was fully functional in P. berghei, indicating functional conservation of GPR180 in Plasmodium spp. With predominant expression and membrane association of GPR180 in sexual stages, GPR180 is a promising target for blocking transmission, and antibodies against GPR180 possess robust transmission-blocking activities.
Collapse
Affiliation(s)
- Peng-peng Wang
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xuefeng Jiang
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Liying Zhu
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Dan Zhou
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Mingyang Hong
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Lu He
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Lumeng Chen
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Shijie Yao
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Yan Zhao
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Guang Chen
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
- Department of Basic Medical Sciences, Taizhou University Hospital, Taizhou University, Taizhou, China
| | - Chengqi Wang
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Liwang Cui
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Yaming Cao
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Xiaotong Zhu
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| |
Collapse
|
8
|
Highly efficient protein expression of Plasmodium vivax surface antigen, Pvs25, by silkworm and its biochemical analysis. Protein Expr Purif 2022; 195-196:106096. [PMID: 35460871 DOI: 10.1016/j.pep.2022.106096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/18/2022] [Accepted: 04/18/2022] [Indexed: 11/20/2022]
Abstract
Plasmodium vivax ookinete surface protein, Pvs25, is a candidate for a transmission-blocking vaccine (TBV) for malaria. Pvs25 has four EGF-like domains containing 22 cysteine residues forming 11 intramolecular disulfide bonds, a structural feature that makes its recombinant protein expression difficult. In this study, we report the high expression of recombinant Pvs25 as a soluble form in silkworm, Bombyx mori. The Pvs25 protein was purified from hemolymphs of larvae and pupae by affinity chromatography. In the Pvs25 expressed by silkworm, no isoforms with inappropriate disulfide bonds were found, requiring no further purification step, which is necessary in the case of Pichia pastoris-based expression systems. The Pvs25 from silkworm was confirmed to be molecularly uniform by sodium dodecyl sulfate gel electrophoresis and size-exclusion chromatography. To examine the immunogenicity, the Pvs25 from B. mori was administered to BALB/c mice subcutaneously with oil adjuvant. The Pvs25 produced by silkworm induced potent and robust immune responses, and the induced antisera correctly recognized P. vivax ookinetes in vitro, demonstrating the potency of Pvs25 from silkworm as a candidate for a malaria TBV. To the best of our knowledge, this is the first study to construct a system for mass-producing malaria TBV antigens using silkworm.
Collapse
|
9
|
Tachibana M, Takashima E, Morita M, Sattabongkot J, Ishino T, Culleton R, Torii M, Tsuboi T. Plasmodium vivax transmission-blocking vaccines: Progress, challenges and innovation. Parasitol Int 2021; 87:102525. [PMID: 34896614 DOI: 10.1016/j.parint.2021.102525] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 12/04/2021] [Indexed: 12/12/2022]
Abstract
Existing control measures have significantly reduced malaria morbidity and mortality in the last two decades, although these reductions are now stalling. Significant efforts have been undertaken to develop malaria vaccines. Recently, extensive progress in malaria vaccine development has been made for Plasmodium falciparum. To date, only the RTS,S/AS01 vaccine has been tested in Phase 3 clinical trials and is now under implementation, despite modest efficacy. Therefore, the development of a malaria transmission-blocking vaccine (TBV) will be essential for malaria elimination. Only a limited number of TBVs have reached pre-clinical or clinical development with several major challenges impeding their development, including low immunogenicity in humans. TBV development efforts against P. vivax, the second major cause of malaria morbidity, lag far behind those for P. falciparum. In this review we summarize the latest progress, challenges and innovations in P. vivax TBV research and discuss how to accelerate its development.
Collapse
Affiliation(s)
- Mayumi Tachibana
- Division of Molecular Parasitology, Proteo-Science Center, Ehime University, Toon, Ehime 791-0295, Japan.
| | - Eizo Takashima
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Ehime 790-8577, Japan.
| | - Masayuki Morita
- 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.
| | - Tomoko Ishino
- Division of Molecular Parasitology, Proteo-Science Center, Ehime University, Toon, Ehime 791-0295, Japan.
| | - Richard Culleton
- Division of Molecular Parasitology, Proteo-Science Center, Ehime University, Toon, Ehime 791-0295, Japan.
| | - Motomi Torii
- Division of Molecular Parasitology, Proteo-Science Center, Ehime University, Toon, Ehime 791-0295, Japan; 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.
| |
Collapse
|
10
|
He W, Baysal C, Lobato Gómez M, Huang X, Alvarez D, Zhu C, Armario‐Najera V, Blanco Perera A, Cerda Bennaser P, Saba‐Mayoral A, Sobrino‐Mengual G, Vargheese A, Abranches R, Alexandra Abreu I, Balamurugan S, Bock R, Buyel JF, da Cunha NB, Daniell H, Faller R, Folgado A, Gowtham I, Häkkinen ST, Kumar S, Sathish Kumar R, Lacorte C, Lomonossoff GP, Luís IM, K.‐C. Ma J, McDonald KA, Murad A, Nandi S, O’Keef B, Parthiban S, Paul MJ, Ponndorf D, Rech E, Rodrigues JC, Ruf S, Schillberg S, Schwestka J, Shah PS, Singh R, Stoger E, Twyman RM, Varghese IP, Vianna GR, Webster G, Wilbers RHP, Christou P, Oksman‐Caldentey K, Capell T. Contributions of the international plant science community to the fight against infectious diseases in humans-part 2: Affordable drugs in edible plants for endemic and re-emerging diseases. PLANT BIOTECHNOLOGY JOURNAL 2021; 19:1921-1936. [PMID: 34181810 PMCID: PMC8486237 DOI: 10.1111/pbi.13658] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/10/2021] [Accepted: 06/22/2021] [Indexed: 05/05/2023]
Abstract
The fight against infectious diseases often focuses on epidemics and pandemics, which demand urgent resources and command attention from the health authorities and media. However, the vast majority of deaths caused by infectious diseases occur in endemic zones, particularly in developing countries, placing a disproportionate burden on underfunded health systems and often requiring international interventions. The provision of vaccines and other biologics is hampered not only by the high cost and limited scalability of traditional manufacturing platforms based on microbial and animal cells, but also by challenges caused by distribution and storage, particularly in regions without a complete cold chain. In this review article, we consider the potential of molecular farming to address the challenges of endemic and re-emerging diseases, focusing on edible plants for the development of oral drugs. Key recent developments in this field include successful clinical trials based on orally delivered dried leaves of Artemisia annua against malarial parasite strains resistant to artemisinin combination therapy, the ability to produce clinical-grade protein drugs in leaves to treat infectious diseases and the long-term storage of protein drugs in dried leaves at ambient temperatures. Recent FDA approval of the first orally delivered protein drug encapsulated in plant cells to treat peanut allergy has opened the door for the development of affordable oral drugs that can be manufactured and distributed in remote areas without cold storage infrastructure and that eliminate the need for expensive purification steps and sterile delivery by injection.
Collapse
Affiliation(s)
- Wenshu He
- Department of Crop and Forest SciencesUniversity of Lleida‐Agrotecnio CERCA CenterLleidaSpain
| | - Can Baysal
- Department of Crop and Forest SciencesUniversity of Lleida‐Agrotecnio CERCA CenterLleidaSpain
| | - Maria Lobato Gómez
- Department of Crop and Forest SciencesUniversity of Lleida‐Agrotecnio CERCA CenterLleidaSpain
| | - Xin Huang
- Department of Crop and Forest SciencesUniversity of Lleida‐Agrotecnio CERCA CenterLleidaSpain
| | - Derry Alvarez
- Department of Crop and Forest SciencesUniversity of Lleida‐Agrotecnio CERCA CenterLleidaSpain
| | - Changfu Zhu
- Department of Crop and Forest SciencesUniversity of Lleida‐Agrotecnio CERCA CenterLleidaSpain
| | - Victoria Armario‐Najera
- Department of Crop and Forest SciencesUniversity of Lleida‐Agrotecnio CERCA CenterLleidaSpain
| | - Aamaya Blanco Perera
- Department of Crop and Forest SciencesUniversity of Lleida‐Agrotecnio CERCA CenterLleidaSpain
| | - Pedro Cerda Bennaser
- Department of Crop and Forest SciencesUniversity of Lleida‐Agrotecnio CERCA CenterLleidaSpain
| | - Andrea Saba‐Mayoral
- Department of Crop and Forest SciencesUniversity of Lleida‐Agrotecnio CERCA CenterLleidaSpain
| | | | - Ashwin Vargheese
- Department of Crop and Forest SciencesUniversity of Lleida‐Agrotecnio CERCA CenterLleidaSpain
| | - Rita Abranches
- Instituto de Tecnologia Química e Biológica António XavierUniversidade Nova de LisboaOeirasPortugal
| | - Isabel Alexandra Abreu
- Instituto de Tecnologia Química e Biológica António XavierUniversidade Nova de LisboaOeirasPortugal
| | - Shanmugaraj Balamurugan
- Plant Genetic Engineering LaboratoryDepartment of BiotechnologyBharathiar UniversityTamil NaduIndia
| | - Ralph Bock
- Max Planck Institute of Molecular Plant PhysiologyPotsdam‐GolmGermany
| | - Johannes F. Buyel
- Fraunhofer Institute for Molecular Biology and Applied Ecology IMEAachenGermany
- Institute for Molecular BiotechnologyRWTH Aachen UniversityAachenGermany
| | - Nicolau B. da Cunha
- Centro de Análise Proteômicas e Bioquímicas de BrasíliaUniversidade Católica de BrasíliaBrasíliaBrazil
| | - Henry Daniell
- School of Dental MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Roland Faller
- Department of Chemical EngineeringUniversity of California, DavisDavisCAUSA
| | - André Folgado
- Instituto de Tecnologia Química e Biológica António XavierUniversidade Nova de LisboaOeirasPortugal
| | - Iyappan Gowtham
- Plant Genetic Engineering LaboratoryDepartment of BiotechnologyBharathiar UniversityTamil NaduIndia
| | - Suvi T. Häkkinen
- Industrial Biotechnology and Food SolutionsVTT Technical Research Centre of Finland LtdEspooFinland
| | - Shashi Kumar
- International Centre for Genetic Engineering and BiotechnologyNew DelhiIndia
| | - Ramalingam Sathish Kumar
- Plant Genetic Engineering LaboratoryDepartment of BiotechnologyBharathiar UniversityTamil NaduIndia
| | - Cristiano Lacorte
- Brazilian Agriculture Research CorporationEmbrapa Genetic Resources and Biotechnology and National Institute of Science and Technology Synthetic in Biology, Parque Estação BiológicaBrasiliaBrazil
| | | | - Ines M. Luís
- Instituto de Tecnologia Química e Biológica António XavierUniversidade Nova de LisboaOeirasPortugal
| | - Julian K.‐C. Ma
- Institute for Infection and ImmunitySt. George’s University of LondonLondonUK
| | - Karen A. McDonald
- Department of Chemical EngineeringUniversity of California, DavisDavisCAUSA
- Global HealthShare InitiativeUniversity of California, DavisDavisCAUSA
| | - Andre Murad
- Brazilian Agriculture Research CorporationEmbrapa Genetic Resources and Biotechnology and National Institute of Science and Technology Synthetic in Biology, Parque Estação BiológicaBrasiliaBrazil
| | - Somen Nandi
- Department of Chemical EngineeringUniversity of California, DavisDavisCAUSA
- Global HealthShare InitiativeUniversity of California, DavisDavisCAUSA
| | - Barry O’Keef
- Division of Cancer Treatment and DiagnosisMolecular Targets ProgramCenter for Cancer ResearchNational Cancer Institute, and Natural Products Branch, Developmental Therapeutics ProgramNational Cancer Institute, NIHFrederickMDUSA
| | - Subramanian Parthiban
- Plant Genetic Engineering LaboratoryDepartment of BiotechnologyBharathiar UniversityTamil NaduIndia
| | - Mathew J. Paul
- Institute for Infection and ImmunitySt. George’s University of LondonLondonUK
| | - Daniel Ponndorf
- Department of Biological ChemistryJohn Innes CentreNorwich Research Park, NorwichUK
| | - Elibio Rech
- Brazilian Agriculture Research CorporationEmbrapa Genetic Resources and Biotechnology and National Institute of Science and Technology Synthetic in Biology, Parque Estação BiológicaBrasiliaBrazil
| | - Julio C.M. Rodrigues
- Brazilian Agriculture Research CorporationEmbrapa Genetic Resources and Biotechnology and National Institute of Science and Technology Synthetic in Biology, Parque Estação BiológicaBrasiliaBrazil
| | - Stephanie Ruf
- Max Planck Institute of Molecular Plant PhysiologyPotsdam‐GolmGermany
| | - Stefan Schillberg
- Fraunhofer Institute for Molecular Biology and Applied Ecology IMEAachenGermany
- Institute for PhytopathologyJustus‐Liebig‐University GiessenGiessenGermany
| | - Jennifer Schwestka
- Institute of Plant Biotechnology and Cell BiologyUniversity of Natural Resources and Life SciencesViennaAustria
| | - Priya S. Shah
- Department of Chemical EngineeringUniversity of California, DavisDavisCAUSA
- Department of Microbiology and Molecular GeneticsUniversity of California, DavisDavisCAUSA
| | - Rahul Singh
- School of Dental MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Eva Stoger
- Institute of Plant Biotechnology and Cell BiologyUniversity of Natural Resources and Life SciencesViennaAustria
| | | | - Inchakalody P. Varghese
- Plant Genetic Engineering LaboratoryDepartment of BiotechnologyBharathiar UniversityTamil NaduIndia
| | - Giovanni R. Vianna
- Brazilian Agriculture Research CorporationEmbrapa Genetic Resources and Biotechnology and National Institute of Science and Technology Synthetic in Biology, Parque Estação BiológicaBrasiliaBrazil
| | - Gina Webster
- Institute for Infection and ImmunitySt. George’s University of LondonLondonUK
| | - Ruud H. P. Wilbers
- Laboratory of NematologyPlant Sciences GroupWageningen University and ResearchWageningenThe Netherlands
| | - Paul Christou
- Department of Crop and Forest SciencesUniversity of Lleida‐Agrotecnio CERCA CenterLleidaSpain
- ICREACatalan Institute for Research and Advanced StudiesBarcelonaSpain
| | | | - Teresa Capell
- Department of Crop and Forest SciencesUniversity of Lleida‐Agrotecnio CERCA CenterLleidaSpain
| |
Collapse
|
11
|
Tripathi AK, Oakley MS, Verma N, Mlambo G, Zheng H, Meredith SM, Essuman E, Puri A, Skelton RA, Takeda K, Majam V, Quakyi IA, Locke E, Morin M, Miura K, Long CA, Kumar S. Plasmodium falciparum Pf77 and male development gene 1 as vaccine antigens that induce potent transmission-reducing antibodies. Sci Transl Med 2021; 13:eabg2112. [PMID: 34108248 PMCID: PMC11018285 DOI: 10.1126/scitranslmed.abg2112] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 04/22/2021] [Indexed: 12/14/2022]
Abstract
Malaria vaccines that disrupt the Plasmodium life cycle in mosquitoes and reduce parasite transmission in endemic areas are termed transmission-blocking vaccines (TBVs). Despite decades of research, there are only a few Plasmodium falciparum antigens that indisputably and reproducibly demonstrate transmission-blocking immunity. So far, only two TBV candidates have advanced to phase 1/2 clinical testing with limited success. By applying an unbiased transcriptomics-based approach, we have identified Pf77 and male development gene 1 (PfMDV-1) as two P. falciparum TBV antigens that, upon immunization, induced antibodies that caused reductions in oocyst counts in Anopheles mosquito midguts in a standard membrane feeding assay. In-depth studies were performed to characterize the genetic diversity of, stage-specific expression by, and natural immunity to these two molecules to evaluate their suitability as TBV candidates. Pf77 and PfMDV-1 display limited antigenic polymorphism, are pan-developmentally expressed within the parasite, and induce naturally occurring antibodies in Ghanaian adults, which raises the prospect of natural boosting of vaccine-induced immune response in endemic regions. Together, these biological properties suggest that Pf77 and PfMDV-1 may warrant further investigation as TBV candidates.
Collapse
Affiliation(s)
- Abhai K Tripathi
- Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Miranda S Oakley
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Nitin Verma
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Godfree Mlambo
- Molecular Microbiology and Immunology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Hong Zheng
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Scott M Meredith
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Edward Essuman
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Ankit Puri
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Richard A Skelton
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Kazuyo Takeda
- Division of Bacterial, Parasitic, and Allergenic Products, Office of Vaccines Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Victoria Majam
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | | | - Emily Locke
- PATH-Malaria Vaccine Initiative, Washington, DC 20001, USA
| | | | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Carole A Long
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Sanjai Kumar
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA.
| |
Collapse
|
12
|
Ramos-Vega A, Monreal-Escalante E, Dumonteil E, Bañuelos-Hernández B, Angulo C. Plant-made vaccines against parasites: bioinspired perspectives to fight against Chagas disease. Expert Rev Vaccines 2021; 20:1373-1388. [PMID: 33612044 DOI: 10.1080/14760584.2021.1893170] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Introduction: Three decades of evidence have demonstrated that plants are an affordable platform for biopharmaceutical production and delivery. For instance, several plant-made recombinant proteins have been approved for commercialization under good manufacturing practice (GMP). Thus far, plant-based vaccine prototypes have been evaluated at pre- and clinical levels. Particularly, plant-made vaccines against parasitic diseases, such as malaria, cysticercosis, and toxoplasmosis have been successfully produced and orally delivered with promising outcomes in terms of immunogenicity and protection. The experience on several approaches and technical strategies over 30 years accounts for their potential low-cost, high scalability, and easy administration.Areas covered: This platform is an open technology to fight against Chagas disease, one of the most important neglected tropical diseases worldwide.Expert opinion: This review provides a perspective for the potential use of plants as a production platform and delivery system of Trypanosoma cruzi recombinant antigens, analyzing the advantages and limitations with respect to plant-made vaccines produced for other parasitic diseases. Plant-made vaccines are envisioned to fight against Chagas disease and other neglected tropical diseases in those countries suffering endemic prevalence.
Collapse
Affiliation(s)
- Abel Ramos-Vega
- Grupo de Inmunología & Vacunología. Centro de Investigaciones Biológicas del Noroeste, S.C. (CIBNOR), Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.c.s. C.p., México
| | - Elizabeth Monreal-Escalante
- Grupo de Inmunología & Vacunología. Centro de Investigaciones Biológicas del Noroeste, S.C. (CIBNOR), Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.c.s. C.p., México.,CONACYT- Centro de Investigaciones Biológicas del Noroeste, S.C. (CIBNOR), Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.c.s. C.p, México
| | - Eric Dumonteil
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, and Vector-Borne and Infectious Disease Research Center, Tulane University, New Orleans, LA, USA
| | - Bernardo Bañuelos-Hernández
- Facultad de Agronomía Y Veterinaria, Universidad de La Salle Bajio, Avenida Universidad 602, Lomas del Campestre, León Guanajuato, México
| | - Carlos Angulo
- Grupo de Inmunología & Vacunología. Centro de Investigaciones Biológicas del Noroeste, S.C. (CIBNOR), Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.c.s. C.p., México
| |
Collapse
|
13
|
Yenkoidiok-Douti L, Canepa GE, Barletta ABF, Barillas-Mury C. In vivo Characterization of Plasmodium berghei P47 (Pbs47) as a Malaria Transmission-Blocking Vaccine Target. Front Microbiol 2020; 11:1496. [PMID: 32719666 PMCID: PMC7348136 DOI: 10.3389/fmicb.2020.01496] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 06/08/2020] [Indexed: 01/08/2023] Open
Abstract
An effective vaccine to reduce malaria transmission is central to control and ultimately achieve disease eradication. Recently, we demonstrated that antibodies targeting the Plasmodium falciparum surface protein P47 (Pfs47) reduce parasite transmission to Anopheles gambiae mosquitoes. Here, Plasmodium berghei (Pb) was used as a model to assess the in vivo efficacy of a P47-targeted transmission blocking vaccine (Pbs47). Mice were immunized following a prime/boost regimen and infected with P. berghei. The effect of immunization on infectivity to mosquitoes was evaluated by direct feeding on P. berghei-infected mice. The key region in Pbs47 where antibody binding confers protection was mapped, and the immunogenicity of this protective antigen was enhanced by conjugation to a virus-like particle. Passive immunization with 100 and 50 μg/mL of anti-Pbs47 IgG reduced oocyst density by 77 and 67%, respectively. Furthermore, affinity purified Pbs47-specific IgG significantly reduced oocyst density by 88 and 77%, respectively at doses as low as 10 and 1 μg/mL. These studies suggest that P47 is a promising transmission blocking target and show that antibodies to the same specific region in Pfs47 and Pbs47 confer protection.
Collapse
Affiliation(s)
- Lampouguin Yenkoidiok-Douti
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Rockville, MD, United States
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
| | - Gaspar E. Canepa
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Rockville, MD, United States
| | - Ana Beatriz F. Barletta
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Rockville, MD, United States
| | - Carolina Barillas-Mury
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Rockville, MD, United States
| |
Collapse
|
14
|
de Jong RM, Tebeje SK, Meerstein‐Kessel L, Tadesse FG, Jore MM, Stone W, Bousema T. Immunity against sexual stage Plasmodium falciparum and Plasmodium vivax parasites. Immunol Rev 2020; 293:190-215. [PMID: 31840844 PMCID: PMC6973022 DOI: 10.1111/imr.12828] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 10/30/2019] [Accepted: 11/14/2019] [Indexed: 12/25/2022]
Abstract
The efficient spread of malaria from infected humans to mosquitoes is a major challenge for malaria elimination initiatives. Gametocytes are the only Plasmodium life stage infectious to mosquitoes. Here, we summarize evidence for naturally acquired anti-gametocyte immunity and the current state of transmission blocking vaccines (TBV). Although gametocytes are intra-erythrocytic when present in infected humans, developing Plasmodium falciparum gametocytes may express proteins on the surface of red blood cells that elicit immune responses in naturally exposed individuals. This immune response may reduce the burden of circulating gametocytes. For both P. falciparum and Plasmodium vivax, there is a solid evidence that antibodies against antigens present on the gametocyte surface, when co-ingested with gametocytes, can influence transmission to mosquitoes. Transmission reducing immunity, reducing the burden of infection in mosquitoes, is a well-acknowledged but poorly quantified phenomenon that forms the basis for the development of TBV. Transmission enhancing immunity, increasing the likelihood or intensity of transmission to mosquitoes, is more speculative in nature but is convincingly demonstrated for P. vivax. With the increased interest in malaria elimination, TBV and monoclonal antibodies have moved to the center stage of malaria vaccine development. Methodologies to prioritize and evaluate products are urgently needed.
Collapse
MESH Headings
- Antibodies, Blocking/immunology
- Antibodies, Protozoan/immunology
- Host-Parasite Interactions/immunology
- Humans
- Immunity
- Immunomodulation
- Life Cycle Stages
- Malaria Vaccines/immunology
- Malaria, Falciparum/immunology
- Malaria, Falciparum/parasitology
- Malaria, Falciparum/prevention & control
- Malaria, Falciparum/transmission
- Malaria, Vivax/immunology
- Malaria, Vivax/parasitology
- Malaria, Vivax/prevention & control
- Malaria, Vivax/transmission
- Plasmodium falciparum/growth & development
- Plasmodium falciparum/immunology
- Plasmodium vivax/growth & development
- Plasmodium vivax/immunology
Collapse
Affiliation(s)
- Roos M. de Jong
- Radboud Institute for Molecular Life SciencesRadboud University Medical CenterNijmegenThe Netherlands
| | | | - Lisette Meerstein‐Kessel
- Radboud Institute for Health SciencesRadboud University Medical CenterNijmegenThe Netherlands
- Centre for Molecular and Biomolecular InformaticsRadboud Institute for Molecular Life SciencesNijmegenThe Netherlands
| | - Fitsum G. Tadesse
- Armauer Hansen Research InstituteAddis AbabaEthiopia
- Radboud Institute for Health SciencesRadboud University Medical CenterNijmegenThe Netherlands
| | - Matthijs M. Jore
- Radboud Institute for Molecular Life SciencesRadboud University Medical CenterNijmegenThe Netherlands
| | - Will Stone
- Department of Immunology and InfectionLondon School of Hygiene and Tropical MedicineLondonUK
| | - Teun Bousema
- Radboud Institute for Health SciencesRadboud University Medical CenterNijmegenThe Netherlands
- Department of Immunology and InfectionLondon School of Hygiene and Tropical MedicineLondonUK
| |
Collapse
|
15
|
Antonelli LR, Junqueira C, Vinetz JM, Golenbock DT, Ferreira MU, Gazzinelli RT. The immunology of Plasmodium vivax malaria. Immunol Rev 2019; 293:163-189. [PMID: 31642531 DOI: 10.1111/imr.12816] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 09/10/2019] [Indexed: 12/13/2022]
Abstract
Plasmodium vivax infection, the predominant cause of malaria in Asia and Latin America, affects ~14 million individuals annually, with considerable adverse effects on wellbeing and socioeconomic development. A clinical hallmark of Plasmodium infection, the paroxysm, is driven by pyrogenic cytokines produced during the immune response. Here, we review studies on the role of specific immune cell types, cognate innate immune receptors, and inflammatory cytokines on parasite control and disease symptoms. This review also summarizes studies on recurrent infections in individuals living in endemic regions as well as asymptomatic infections, a serious barrier to eliminating this disease. We propose potential mechanisms behind these repeated and subclinical infections, such as poor induction of immunological memory cells and inefficient T effector cells. We address the role of antibody-mediated resistance to P. vivax infection and discuss current progress in vaccine development. Finally, we review immunoregulatory mechanisms, such as inhibitory receptors, T regulatory cells, and the anti-inflammatory cytokine, IL-10, that antagonizes both innate and acquired immune responses, interfering with the development of protective immunity and parasite clearance. These studies provide new insights for the clinical management of symptomatic as well as asymptomatic individuals and the development of an efficacious vaccine for vivax malaria.
Collapse
Affiliation(s)
- Lis R Antonelli
- Instituto de Pesquisas Rene Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil
| | - Caroline Junqueira
- Instituto de Pesquisas Rene Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil
| | - Joseph M Vinetz
- Section of Infectious Diseases, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Douglas T Golenbock
- Division of Infectious Disease and immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Marcelo U Ferreira
- Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Ricardo T Gazzinelli
- Instituto de Pesquisas Rene Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Brazil.,Division of Infectious Disease and immunology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA.,Plataforma de Medicina Translacional, Fundação Oswaldo Cruz, Ribeirão Preto, Brazil
| |
Collapse
|
16
|
Cheng CW, Jongwutiwes S, Putaporntip C, Jackson AP. Clinical expression and antigenic profiles of a Plasmodium vivax vaccine candidate: merozoite surface protein 7 (PvMSP-7). Malar J 2019; 18:197. [PMID: 31196098 PMCID: PMC6567670 DOI: 10.1186/s12936-019-2826-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 06/04/2019] [Indexed: 12/18/2022] Open
Abstract
Background Vivax malaria is the predominant form of malaria outside Africa, affecting about 14 million people worldwide, with about 2.5 billion people exposed. Development of a Plasmodium vivax vaccine is a priority, and merozoite surface protein 7 (MSP-7) has been proposed as a plausible candidate. The P. vivax genome contains 12 MSP-7 genes, which contribute to erythrocyte invasion during blood-stage infection. Previous analysis of MSP-7 sequence diversity suggested that not all paralogs are functionally equivalent. To explore MSP-7 functional diversity, and to identify the best vaccine candidate within the family, MSP-7 expression and antigenicity during bloodstream infections were examined directly from clinical isolates. Methods Merozoite surface protein 7 gene expression was profiled using RNA-seq data from blood samples isolated from ten human patients with vivax malaria. Differential expression analysis and co-expression cluster analysis were used to relate PvMSP-7 expression to genetic markers of life cycle stage. Plasma from vivax malaria patients was also assayed using a custom peptide microarray to measure antibody responses against the coding regions of 12 MSP-7 paralogs. Results Ten patients presented diverse transcriptional profiles that comprised four patient groups. Two MSP-7 paralogs, 7A and 7F, were expressed abundantly in all patients, while other MSP-7 genes were uniformly rare (e.g. 7J). MSP-7H and 7I were significantly more abundant in patient group 4 only, (two patients having experienced longer patency), and were co-expressed with a schizont-stage marker, while negatively associated with liver-stage and gametocyte-stage markers. Screening infections with a PvMSP-7 peptide array identified 13 linear B-cell epitopes in five MSP-7 paralogs that were recognized by plasma from all patients. Conclusions These results show that MSP-7 family members vary in expression profile during blood infections; MSP-7A and 7F are expressed throughout the intraerythrocytic development cycle, while expression of other paralogs is focused on the schizont. This may reflect developmental regulation, and potentially functional differentiation, within the gene family. The frequency of B-cell epitopes among paralogs also varies, with MSP-7A and 7L consistently the most immunogenic. Thus, MSP-7 paralogs cannot be assumed to have equal potential as vaccines. This analysis of clinical infections indicates that the most abundant and immunogenic paralog is MSP-7A. Electronic supplementary material The online version of this article (10.1186/s12936-019-2826-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Chew Weng Cheng
- Department of Infection Biology, Institute of Infection and Global Health, University of Liverpool, 146 Brownlow Hill, Liverpool, L3 5RF, UK.,Molecular Biology of Malaria and Opportunistic Parasites Research Unit, Department of Parasitology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Somchai Jongwutiwes
- Molecular Biology of Malaria and Opportunistic Parasites Research Unit, Department of Parasitology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Chaturong Putaporntip
- Molecular Biology of Malaria and Opportunistic Parasites Research Unit, Department of Parasitology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Andrew P Jackson
- Department of Infection Biology, Institute of Infection and Global Health, University of Liverpool, 146 Brownlow Hill, Liverpool, L3 5RF, UK.
| |
Collapse
|
17
|
McCaffery JN, Fonseca JA, Singh B, Cabrera-Mora M, Bohannon C, Jacob J, Arévalo-Herrera M, Moreno A. A Multi-Stage Plasmodium vivax Malaria Vaccine Candidate Able to Induce Long-Lived Antibody Responses Against Blood Stage Parasites and Robust Transmission-Blocking Activity. Front Cell Infect Microbiol 2019; 9:135. [PMID: 31119106 PMCID: PMC6504793 DOI: 10.3389/fcimb.2019.00135] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 04/15/2019] [Indexed: 12/13/2022] Open
Abstract
Malaria control and interventions including long-lasting insecticide-treated nets, indoor residual spraying, and intermittent preventative treatment in pregnancy have resulted in a significant reduction in the number of Plasmodium falciparum cases. Considerable efforts have been devoted to P. falciparum vaccines development with much less to P. vivax. Transmission-blocking vaccines, which can elicit antibodies targeting Plasmodium antigens expressed during sexual stage development and interrupt transmission, offer an alternative strategy to achieve malaria control. The post-fertilization antigen P25 mediates several functions essential to ookinete survival but is poorly immunogenic in humans. Previous clinical trials targeting this antigen have suggested that conjugation to a carrier protein could improve the immunogenicity of P25. Here we report the production, and characterization of a vaccine candidate composed of a chimeric P. vivax Merozoite Surface Protein 1 (cPvMSP1) genetically fused to P. vivax P25 (Pvs25) designed to enhance CD4+ T cell responses and its assessment in a murine model. We demonstrate that antibodies elicited by immunization with this chimeric protein recognize both the erythrocytic and sexual stages and are able to block the transmission of P. vivax field isolates in direct membrane-feeding assays. These findings provide support for the continued development of multi-stage transmission blocking vaccines targeting the life-cycle stage responsible for clinical disease and the sexual-stage development accountable for disease transmission simultaneously.
Collapse
Affiliation(s)
- Jessica N. McCaffery
- Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
| | - Jairo A. Fonseca
- Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
- Division of Infectious Diseases, Department of Medicine, Emory University, Atlanta, GA, United States
| | - Balwan Singh
- Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
| | - Monica Cabrera-Mora
- Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
| | - Caitlin Bohannon
- Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
| | - Joshy Jacob
- Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, United States
| | - Myriam Arévalo-Herrera
- Caucaseco Scientific Research Center, Malaria Vaccine and Drug Development Center, Cali, Colombia
| | - Alberto Moreno
- Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
- Division of Infectious Diseases, Department of Medicine, Emory University, Atlanta, GA, United States
| |
Collapse
|
18
|
Sala KA, Angrisano F, Da DF, Taylor IJ, Churcher TS, Blagborough AM. Immunization with Transgenic Rodent Malaria Parasites Expressing Pfs25 Induces Potent Transmission-Blocking Activity. Sci Rep 2018; 8:1573. [PMID: 29371619 PMCID: PMC5785477 DOI: 10.1038/s41598-017-18831-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 12/14/2017] [Indexed: 12/14/2022] Open
Abstract
An anti-malarial transmission blocking vaccine (TBV) would be an important tool for disease control or elimination, though current candidates have failed to induce high efficacy in clinical studies. The ookinete surface protein P25 is a primary target for TBV development, but heterologous expression of P25 with appropriate conformation is problematic and a pre-requisite for achieving functional titers. A potential alternative to recombinant/sub-unit vaccine is immunization with a non-pathogenic, whole-parasite vaccine. This study examines the ability of a purified transgenic rodent-malaria parasite (PbPfs25DR3), expressing Plasmodium falciparum P25 in native conformation on the P. berghei ookinete surface, to act as a TBV. Vaccination with purified PbPfs25DR3 ookinetes produces a potent anti-Pfs25 response and high transmission-blocking efficacy in the laboratory, findings that are then translated to experimentation on natural field isolates of P. falciparum from infected individuals in Burkina Faso. Efficacy is demonstrated in the lab and the field (up to 93.3%/97.1% reductions in transmission intensity respectively), with both a homologous strategy with one and two boosts, and as part of a prime-boost regime, providing support for the future development of a whole-parasite TBV.
Collapse
Affiliation(s)
- K A Sala
- Department of Life Sciences, Sir Alexander Fleming Building, Imperial College London, Imperial College Road, South Kensington, London, SW7 2AZ, UK
| | - F Angrisano
- Department of Life Sciences, Sir Alexander Fleming Building, Imperial College London, Imperial College Road, South Kensington, London, SW7 2AZ, UK
| | - D F Da
- Institut de Recherche en Sciences de la Santé, 399 Avenue de la Liberté, BP 545, Bobo-Dioulasso, Burkina Faso
| | - I J Taylor
- Jenner Institute, The University of Oxford, Roosevelt Road, Oxford, OX9 2PP, UK
| | - T S Churcher
- MRC Centre for Outbreak Analysis and Modelling, Department of Infectious Disease Epidemiology, Imperial College London, Norfolk Place, London, W2 1PG, UK
| | - A M Blagborough
- Department of Life Sciences, Sir Alexander Fleming Building, Imperial College London, Imperial College Road, South Kensington, London, SW7 2AZ, UK.
| |
Collapse
|
19
|
Othman AS, Marin-Mogollon C, Salman AM, Franke-Fayard BM, Janse CJ, Khan SM. The use of transgenic parasites in malaria vaccine research. Expert Rev Vaccines 2017; 16:1-13. [DOI: 10.1080/14760584.2017.1333426] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Ahmad Syibli Othman
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
- Faculty of Health Sciences, Universiti Sultan Zainal Abidin, Terengganu, Malaysia
| | - Catherin Marin-Mogollon
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | | | - Blandine M. Franke-Fayard
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | - Chris J. Janse
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | - Shahid M. Khan
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| |
Collapse
|
20
|
López C, Yepes-Pérez Y, Hincapié-Escobar N, Díaz-Arévalo D, Patarroyo MA. What Is Known about the Immune Response Induced by Plasmodium vivax Malaria Vaccine Candidates? Front Immunol 2017; 8:126. [PMID: 28243235 PMCID: PMC5304258 DOI: 10.3389/fimmu.2017.00126] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Accepted: 01/25/2017] [Indexed: 12/15/2022] Open
Abstract
Malaria caused by Plasmodium vivax continues being one of the most important infectious diseases around the world; P. vivax is the second most prevalent species and has the greatest geographic distribution. Developing an effective antimalarial vaccine is considered a relevant control strategy in the search for means of preventing the disease. Studying parasite-expressed proteins, which are essential in host cell invasion, has led to identifying the regions recognized by individuals who are naturally exposed to infection. Furthermore, immunogenicity studies have revealed that such regions can trigger a robust immune response that can inhibit sporozoite (hepatic stage) or merozoite (erythrocyte stage) invasion of a host cell and induce protection. This review provides a synthesis of the most important studies to date concerning the antigenicity and immunogenicity of both synthetic peptide and recombinant protein candidates for a vaccine against malaria produced by P. vivax.
Collapse
Affiliation(s)
- Carolina López
- Molecular Biology and Immunology Department, Fundación Instituto de Immunología de Colombia (FIDIC), Bogotá, Colombia; PhD Programme in Biomedical and Biological Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Yoelis Yepes-Pérez
- Molecular Biology and Immunology Department, Fundación Instituto de Immunología de Colombia (FIDIC), Bogotá, Colombia; MSc Programme in Microbiology, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Natalia Hincapié-Escobar
- Molecular Biology and Immunology Department, Fundación Instituto de Immunología de Colombia (FIDIC) , Bogotá , Colombia
| | - Diana Díaz-Arévalo
- Molecular Biology and Immunology Department, Fundación Instituto de Immunología de Colombia (FIDIC), Bogotá, Colombia; Universidad de Ciencias Aplicadas y Ambientales (UDCA), Bogotá, Colombia
| | - Manuel A Patarroyo
- Molecular Biology and Immunology Department, Fundación Instituto de Immunología de Colombia (FIDIC), Bogotá, Colombia; Basic Sciences Department, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| |
Collapse
|
21
|
Zheng W, Liu F, He Y, Liu Q, Humphreys GB, Tsuboi T, Fan Q, Luo E, Cao Y, Cui L. Functional characterization of Plasmodium berghei PSOP25 during ookinete development and as a malaria transmission-blocking vaccine candidate. Parasit Vectors 2017; 10:8. [PMID: 28057055 PMCID: PMC5217559 DOI: 10.1186/s13071-016-1932-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 12/06/2016] [Indexed: 12/23/2022] Open
Abstract
Background Plasmodium ookinete surface proteins as post-fertilization target antigens are potential malaria transmission-blocking vaccine (TBV) candidates. Putative secreted ookinete protein 25 (PSOP25) is a highly conserved ookinete surface protein, and has been shown to be a promising novel TBV target. Here, we further investigated the TBV activities of the full-length recombinant PSOP25 (rPSOP25) protein in Plasmodium berghei, and characterized the potential functions of PSOP25 during the P. berghei life-cycle. Methods We expressed the full-length P. berghei PSOP25 protein in a prokaryotic expression system, and developed polyclonal mouse antisera and a monoclonal antibody (mAb) against the recombinant protein. Indirect immunofluorescence assay (IFA) and Western blot were used to test the specificity of antibodies. The transmission-blocking (TB) activities of antibodies were evaluated by the in vitro ookinete conversion assay and by direct mosquito feeding assay (DFA). Finally, the function of PSOP25 during Plasmodium development was studied by deleting the psop25 gene. Results Both polyclonal mouse antisera and anti-rPSOP25 mAb recognized the PSOP25 proteins in the parasites, and IFA showed the preferential expression of PSOP25 on the surface of zygotes, retorts and mature ookinetes. In vitro, these antibodies significantly inhibited ookinetes formation in an antibody concentration-dependent manner. In DFA, mice immunized with the rPSOP25 and those receiving passive transfer of the anti-rPSOP25 mAb reduced the prevalence of mosquito infection by 31.2 and 26.1%, and oocyst density by 66.3 and 63.3%, respectively. Genetic knockout of the psop25 gene did not have a detectable impact on the asexual growth of P. berghei, but significantly affected the maturation of ookinetes and the formation of midgut oocysts. Conclusions The full-length rPSOP25 could elicit strong antibody response in mice. Polyclonal and monoclonal antibodies against PSOP25 could effectively block the formation of ookinetes in vitro and transmission of the parasites to mosquitoes. Genetic manipulation study indicated that PSOP25 is required for ookinete maturation in P. berghei. These results support further testing of the PSOP25 orthologs in human malaria parasites as promising TBV candidates. Electronic supplementary material The online version of this article (doi:10.1186/s13071-016-1932-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wenqi Zheng
- Department of Pathogen Biology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, 110001, China.,Laboratory of Surgery, The Affiliated Hospital, Inner Mongolia Medical University, Hohhot, 010050, China
| | - Fei Liu
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, 110001, China
| | - Yiwen He
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, 110001, China
| | - Qingyang Liu
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, 110001, China
| | - Gregory B Humphreys
- Department of Entomology, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Takafumi Tsuboi
- Cell-free Science and Technology Research Center, Ehime University, Matsuyama, Ehime, 790-8577, Japan
| | - Qi Fan
- Dalian Institute of Biotechnology, Dalian, Liaoning, China
| | - Enjie Luo
- Department of Pathogen Biology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, 110001, China
| | - Yaming Cao
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, 110001, China.
| | - Liwang Cui
- Department of Entomology, The Pennsylvania State University, University Park, PA, 16802, USA
| |
Collapse
|