1
|
Song Y, Huang WC, Ivanochko D, Long C, Li Q, Zhou L, Julien JP, Miura K, Lovell JF. 50-Fold Adjuvant and 20-Fold Antigen Vaccine Dose Sparing from Nanoliposome Display of a Stabilized Malarial Protein Antigen. ACS NANO 2025; 19:10103-10112. [PMID: 40099532 DOI: 10.1021/acsnano.4c16865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Displaying soluble vaccine protein antigens onto the surface of adjuvanted nanoliposomes can enhance the magnitude of elicited antibody responses. In this study, we examine this approach with respect to dose sparing, for not only the antigen component but also the adjuvant dose in the vaccine. Using a structurally stabilized Pfs48/45 derived malarial protein as a model antigen, we confirmed the protein rapidly displayed on the surface of immunogenic liposomes containing cobalt porphyrin phospholipid (CoPoP; for antigen display via His-tag interaction) along with the immunostimulatory adjuvants monophosphoryl lipid A (MPLA) and QS-21. Mice were immunized with a fixed protein antigen dose with varying adjuvant doses to estimate the extent of adjuvant sparing. In mice vaccinated at a fixed protein antigen dose, liposome-bound Pfs48/45 achieved superior antibody IgG titers compared to the soluble (nonbound) form at all assessed adjuvant doses, reflecting MPLA and QS-21 adjuvant dose sparing of at least 50-fold. The primary driver of adjuvant sparing in these conditions was presentation of the antigen in a nanoparticle format, and potent responses were achieved even without co-delivery of antigen and adjuvant within the same particle, provided that adjuvant and liposome-displayed antigen were co-administered to the same injection site. By keeping the adjuvant dose fixed and varying the antigen dose in a comparable experimental design, ∼20-fold antigen dose sparing was observed with liposome display. This case study illustrates the potential of antigen-display nanotechnologies, such as CoPoP nanoliposomes, to achieve substantial adjuvant and antigen dose sparing, which could theoretically facilitate the deployment of future vaccines.
Collapse
Affiliation(s)
- Yiting Song
- Department of Biomedical Engineering, State University of New York at Buffalo, Buffalo, New York 14260, United States
| | - Wei-Chiao Huang
- Department of Biomedical Engineering, State University of New York at Buffalo, Buffalo, New York 14260, United States
| | - Danton Ivanochko
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, Ontario M5G 0A4, Canada
| | - Carole Long
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland 20852, United States
| | - Qinzhe Li
- Department of Biomedical Engineering, State University of New York at Buffalo, Buffalo, New York 14260, United States
| | - Luwen Zhou
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland 20852, United States
| | - Jean-Philippe Julien
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, Ontario M5G 0A4, Canada
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland 20852, United States
| | - Jonathan F Lovell
- Department of Biomedical Engineering, State University of New York at Buffalo, Buffalo, New York 14260, United States
| |
Collapse
|
2
|
Adeleke MA. Computational Development of Transmission-Blocking Vaccine Candidates Based on Fused Antigens of Pre- and Post-fertilization Gametocytes Against Plasmodium falciparum. Bioinform Biol Insights 2025; 19:11779322241306215. [PMID: 40034580 PMCID: PMC11873872 DOI: 10.1177/11779322241306215] [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: 05/01/2024] [Accepted: 11/21/2024] [Indexed: 03/05/2025] Open
Abstract
Plasmodium falciparum is the most fatal species of malaria parasites in humans. Attempts at developing vaccines against the malaria parasites have not been very successful even after the approval of the RTS, S/AS01 vaccine. There is a continuous need for more effective vaccines including sexual-stage antigens that could block the transmission of malaria parasites between mosquitoes and humans. Low immunogenicity, expression, and stability are some of the challenges of transmission-blocking vaccine (TBV). This study was designed to computationally identify TBV candidates based on fused antigens by combining highly antigenic peptides from prefertilization (Pfs230, Pfs48/45) and postfertilization (Pfs25, Pfs28) gametocytes. The peptides were selected based on their antigenicity, nonallergenicity, and lack of similarity with the human proteome. Two fused antigens vaccine candidates (FAVCs) were constructed using Flagellin Salmonella enterica (FAVC-FSE) and Cholera toxin B (FAVC-CTB) as adjuvants. The constructs were evaluated for their physicochemical properties, structural stability, immunogenicity, and potential to elicit cross-protection across multiple Plasmodium species. The results yielded antigenic peptides, with antigenicity scores between 0.7589 and 1.1821. The structural analysis of FAVC-FSE and FAVC-CTB showed a Z-score of -6.70 and -4.79, a Ramachandran plot of 96.94% and 94.86% with overall quality of 94.20% and 89.85%, respectively. The FAVCs contained CD8+, CD4+, and linear B-cell epitopes with antigenicity scores between 1.2089 and 2.8623, 0.5663 and 2.4132, and 1.5196 and 2.2212, respectively. Each FAVC generated 6 conformational B-cells. High population coverage values were recorded for the FAVCs. The ability of the FAVCs to trigger immune response was evaluated through an in silico immune stimulation. The low-binding interaction energy that resulted from molecular docking and dynamics simulations showed a strong affinity of FAVCs to Toll-like receptor 5 (TLR5). The results indicate that the FAVC-FSE vaccine candidate is more promising to interrupt P falciparum transmission and provides a baseline for experimental validation.
Collapse
Affiliation(s)
- Matthew A. Adeleke
- Discipline of Genetics, School of Life Sciences, College of Agriculture, Engineering and Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
3
|
Kuesap J, Suphakhonchuwong N, Eksonthi B, Huaihongthong S. Genetic polymorphisms of Plasmodium vivax transmission-blocking vaccine candidates Pvs48/45 and Pvs47 in Thailand. Malar J 2025; 24:63. [PMID: 40016697 PMCID: PMC11866859 DOI: 10.1186/s12936-025-05305-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: 08/25/2024] [Accepted: 02/22/2025] [Indexed: 03/01/2025] Open
Abstract
BACKGROUND The genetic diversity of malaria parasites varies between regions in the world. The genetic polymorphisms of the genes Pvs48/45 and Pvs47 which encode gametocyte/gamete proteins of Plasmodium vivax, were studied because of their potential as transmission-blocking vaccine (TBV) targets. The aim of the present study was to investigate the genetic diversity of Pvs48/45 and Pvs47 in clinical isolates from endemic areas of Thailand. METHODS Plasmodium vivax samples collected from four provinces neighbouring either Myanmar or Malaysia were analysed using polymerase chain reaction and nucleotide sequencing. RESULTS: Fifteen and 18 amino acid substitutions were observed in 36 Pvs48/45 and 62 Pvs47 deduced amino acid sequences, respectively. Eleven haplotypes were identified in Pvs48/45 and 26 in Pvs47. Overall, low nucleotide diversities were observed for Pvs48/45 (π = 0.00104) and Pvs47 (π = 0.00321). Tajima's D, and Fu and Li's D* and F* values were negative for both genes, Pvs48/45 and Pvs47 while a significant difference was found in Pvs48/45 (P < 0.05). CONCLUSION The limited polymorphism of the two investigated TBV candidate antigens observed in this study is consistent with findings in worldwide isolates. The collected genetic diversity data could be helpful for developing effective TBVs in malaria-endemic areas.
Collapse
Affiliation(s)
- Jiraporn Kuesap
- Faculty of Allied Health Sciences, Thammasat University, 99 Moo 18 Khlong Nueng, Khlong Luang, Pathumthani, 12120, Thailand.
| | - Nutnicha Suphakhonchuwong
- Faculty of Allied Health Sciences, Thammasat University, 99 Moo 18 Khlong Nueng, Khlong Luang, Pathumthani, 12120, Thailand
- Faculty of Medical Technology, Rangsit University, Pathumthani, Thailand
| | - Benyapa Eksonthi
- Faculty of Allied Health Sciences, Thammasat University, 99 Moo 18 Khlong Nueng, Khlong Luang, Pathumthani, 12120, Thailand
| | - Saranchana Huaihongthong
- Faculty of Allied Health Sciences, Thammasat University, 99 Moo 18 Khlong Nueng, Khlong Luang, Pathumthani, 12120, Thailand
| |
Collapse
|
4
|
Cao Y, Hayashi CTH, Araujo MDS, Tripathi AK, Andrade AO, Medeiros JF, Vinetz J, Kumar N. Evaluation of combination vaccines targeting transmission of Plasmodium falciparum and P. vivax. Vaccine 2024; 42:126140. [PMID: 39033079 PMCID: PMC11338703 DOI: 10.1016/j.vaccine.2024.07.041] [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: 04/18/2024] [Revised: 07/02/2024] [Accepted: 07/12/2024] [Indexed: 07/23/2024]
Abstract
Transmission-blocking vaccines interrupting malaria transmission within mosquitoes represent an ideal public health tool to eliminate malaria at the population level. Plasmodium falciparum and P. vivax account for more than 90% of the global malaria burden, co-endemic in many regions of the world. P25 and P48/45 are two leading candidates for both species and have shown promising transmission-blocking activity in preclinical and clinical studies. However, neither of these target antigens as individual vaccines has induced complete transmission inhibition in mosquitoes. In this study, we assessed immunogenicity of combination vaccines based on P25 and P48/45 using a DNA vaccine platform to broaden vaccine specificity against P. falciparum and P. vivax. Individual DNA vaccines encoding Pvs25, Pfs25, Pvs48/45 and Pfs48/45, as well as various combinations including (Pvs25 + Pvs48/45), (Pfs25 + Pfs48/45), (Pvs25 + Pfs25), and (Pvs48/45 + Pfs48/45), were evaluated in mice using in vivo electroporation. Potent antibody responses were induced in mice immunized with individual and combination DNA vaccines, and specific antibody responses were not compromised when combinations of DNA vaccines were evaluated against individual DNA vaccines. The anti-Pvs25 IgG from individual and combination groups revealed concentration-dependent transmission-reducing activity (TRA) in direct membrane feeding assays (DMFA) using blood from P. vivax-infected donors in Brazil and independently in ex vivo MFA using Pvs25-transgenic P. berghei. Similarly, anti-Pfs25 and anti-Pfs48/45 IgGs from mice immunized with Pfs25 and Pfs48/45 DNA vaccines individually and in various combinations revealed antibody dose-dependent TRA in standard membrane feeding assays (SMFA) using culture-derived P. falciparum gametocytes. However, antibodies induced by immunization with Pvs48/45 DNA vaccines were ineffective in DMFA and require further vaccine construct optimization, considering the possibility of induction of both transmission-blocking and transmission-enhancing antibodies revealed by competition ELISA. These studies provide a rationale for combining multiple antigens to simultaneously target transmission of malaria caused by P. falciparum and P. vivax.
Collapse
MESH Headings
- Malaria Vaccines/immunology
- Malaria Vaccines/administration & dosage
- Animals
- Malaria, Falciparum/prevention & control
- Malaria, Falciparum/transmission
- Malaria, Falciparum/immunology
- Plasmodium falciparum/immunology
- Plasmodium falciparum/genetics
- Plasmodium vivax/immunology
- Plasmodium vivax/genetics
- Malaria, Vivax/prevention & control
- Malaria, Vivax/transmission
- Malaria, Vivax/immunology
- Mice
- Vaccines, DNA/immunology
- Vaccines, DNA/administration & dosage
- Antibodies, Protozoan/immunology
- Antibodies, Protozoan/blood
- Female
- Vaccines, Combined/immunology
- Vaccines, Combined/administration & dosage
- Antigens, Protozoan/immunology
- Antigens, Protozoan/genetics
- Protozoan Proteins/immunology
- Protozoan Proteins/genetics
- Mice, Inbred BALB C
- Humans
Collapse
Affiliation(s)
- Yi Cao
- Department of Global Health, Milken Institute School of Public Health, George Washington University, Washington D.C., USA
| | - Clifford T H Hayashi
- Department of Global Health, Milken Institute School of Public Health, George Washington University, Washington D.C., USA
| | - Maisa da Silva Araujo
- Plataforma de Produção e Infecção de Vetores da Malária, Laboratório de Entomologia - FIOCRUZ RO, Rua da Beira 7671, CEP 76812-245 Porto Velho RO, Brazil; Programa de Pós-Graduação em Saúde Pública, Faculdade de Saúde Pública, Universidade Federal de São Paulo, São Paulo 01246-904, SP, Brazil
| | - Abhai K Tripathi
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Alice Oliveira Andrade
- Plataforma de Produção e Infecção de Vetores da Malária, Laboratório de Entomologia - FIOCRUZ RO, Rua da Beira 7671, CEP 76812-245 Porto Velho RO, Brazil; Programa de Pós-Graduação em Saúde Pública, Faculdade de Saúde Pública, Universidade Federal de São Paulo, São Paulo 01246-904, SP, Brazil
| | - Jansen Fernandes Medeiros
- Plataforma de Produção e Infecção de Vetores da Malária, Laboratório de Entomologia - FIOCRUZ RO, Rua da Beira 7671, CEP 76812-245 Porto Velho RO, Brazil; Programa de Pós-Graduação em Biologia Experimental, Fundação Universidade Federal de Rondônia, Fiocruz Rondônia 76812-245, Brazil
| | - Joseph Vinetz
- Section of Infectious Diseases, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA; Department of Cellular and Molecular Sciences, Faculty of Sciences, and Alexander von Humboldt Institute of Tropical Medicine, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Nirbhay Kumar
- Department of Global Health, Milken Institute School of Public Health, George Washington University, Washington D.C., USA.
| |
Collapse
|
5
|
Tajudeen YA, Oladipo HJ, Yusuff SI, Abimbola SO, Abdulkadir M, Oladunjoye IO, Omotosho AO, Egbewande OM, Shittu HD, Yusuf RO, Ogundipe O, Muili AO, Afolabi AO, Dahesh SMA, Gameil MAM, El-Sherbini MS. A landscape review of malaria vaccine candidates in the pipeline. Trop Dis Travel Med Vaccines 2024; 10:19. [PMID: 39085983 PMCID: PMC11293096 DOI: 10.1186/s40794-024-00222-3] [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: 08/21/2023] [Accepted: 04/15/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND Globally, malaria continues to pose a major health challenge, with approximately 247 million cases of the illness and 627,000 deaths reported in 2021. However, the threat is particularly pronounced in sub-Saharan African countries, where pregnant women and children under the age of five face heightened vulnerability to the disease. As a result, the imperative to develop malaria vaccines especially for these vulnerable populations, remains crucial in the pursuit of malaria eradication. However, despite decades of research, effective vaccine development faces technical challenges, including the rapid spread of drug-resistant parasite strains, the complex parasite lifecycle, the development of liver hypnozoites with potential for relapse, and evasion of the host immune system. This review aims to discuss the different malaria vaccine candidates in the pipeline, highlighting different approaches used for adjuvating these candidates, their benefits, and outcomes, and summarizing the progress of these vaccine candidates under development. METHOD A comprehensive web-based search for peer-reviewed journal articles published in SCOPUS, MEDLINE (via PubMed), Science Direct, WHO, and Advanced Google Scholar databases was conducted from 1990 to May 2022. Context-specific keywords such as "Malaria", "Malaria Vaccine", "Malaria Vaccine Candidates", "Vaccine Development", "Vaccine Safety", "Clinical Trials", "mRNA Vaccines", "Viral Vector Vaccines", "Protein-based Vaccines", "Subunit Vaccines", "Vaccine Adjuvants", "Vaccine-induced Immune Responses", and "Immunogenicity" were emphatically considered. Articles not directly related to malaria vaccine candidates in preclinical and clinical stages of development were excluded. RESULTS Various approaches have been studied for malaria vaccine development, targeting different parasite lifecycle stages, including the pre-erythrocytic, erythrocytic, and sexual stages. The RTS, S/AS01 vaccine, the first human parasite vaccine reaching WHO-listed authority maturity level 4, has demonstrated efficacy in preventing clinical malaria in African children. However, progress was slow in introducing other safe, and feasible malaria vaccines through clinical trials . Recent studies highlight the potential effectiveness of combining pre-erythrocytic and blood-stage vaccines, along with the advantages of mRNA vaccines for prophylaxis and treatment, and nonstructural vaccines for large-scale production. CONCLUSION Malaria vaccine candidates targeting different lifecycle stages of the parasite range from chemoprophylaxis vaccination to cross-species immune protection. The use of a multi-antigen, multi-stage combinational vaccine is therefore essential in the context of global health. This demands careful understanding and critical consideration of the long-term multi-faceted interplay of immune interference, co-dominance, complementary immune response, molecular targets, and adjuvants affecting the overall vaccine-induced immune response. Despite challenges, advancements in clinical trials and vaccination technology offer promising possibilities for novel approaches in malaria vaccine development.
Collapse
Affiliation(s)
- Yusuf Amuda Tajudeen
- Department of Microbiology, Faculty of Life Sciences, University of Ilorin, P.M.B. 1515, Ilorin, 240003, Nigeria
- Department of Epidemiology and Medical Statistics, Faculty of Public Health, College of Medicine, University of Ibadan, P.M.B 5017 G.P.O, Ibadan, Oyo State, Nigeria
| | - Habeebullah Jayeola Oladipo
- Department of Microbiology, Faculty of Life Sciences, University of Ilorin, P.M.B. 1515, Ilorin, 240003, Nigeria
- Faculty of Pharmaceutical Sciences, University of Ilorin, P.M.B. 1515, Ilorin, 240003, Nigeria
| | - Sodiq Inaolaji Yusuff
- Department of Medicine, Faculty of Clinical Sciences, Obafemi Awolowo University, Ibadan- Ife Rd, Ife, 220282, Osun State, Nigeria
| | - Samuel O Abimbola
- Cyprus International Institute of Environmental and Public Health, Cyprus University of Technology, Limassol, 3036, Cyprus
| | - Muritala Abdulkadir
- Faculty of Pharmaceutical Sciences, University of Ilorin, P.M.B. 1515, Ilorin, 240003, Nigeria
| | - Iyiola Olatunji Oladunjoye
- Department of Microbiology, Faculty of Life Sciences, University of Ilorin, P.M.B. 1515, Ilorin, 240003, Nigeria
| | - Abass Olawale Omotosho
- Department of Microbiology, Faculty of Pure and Applied Sciences, Kwara State University, P.M.B 1530, Malete-Ilorin, Ilorin, Nigeria
| | | | | | - Rashidat Onyinoyi Yusuf
- Faculty of Pharmaceutical Sciences, University of Ilorin, P.M.B. 1515, Ilorin, 240003, Nigeria
| | - Oluwatosin Ogundipe
- Department of Epidemiology and Medical Statistics, Faculty of Public Health, College of Medicine, University of Ibadan, P.M.B 5017 G.P.O, Ibadan, Oyo State, Nigeria
| | - Abdulbasit Opeyemi Muili
- Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology, P.M.B 4000, Ogbomosho, Oyo State, Nigeria
| | - Abdullateef Opeyemi Afolabi
- Faculty of Biomedical Sciences, Department of Microbiology and Immunology, Kampala International University, Bushenyi, Uganda.
| | - Salwa M A Dahesh
- Research Institute of Medical Entomology, General Organization for Teaching Hospitals and Institutes, GOTHI, Damietta, Egypt
| | | | - Mona Said El-Sherbini
- Department of Medical Parasitology, Faculty of Medicine, Cairo University, Cairo, 11562, Egypt
| |
Collapse
|
6
|
Bansal GP, Araujo MDS, Cao Y, Shaffer E, Araujo JE, Medeiros JF, Hayashi C, Vinetz J, Kumar N. Transmission-reducing and -enhancing monoclonal antibodies against Plasmodium vivax gamete surface protein Pvs48/45. Infect Immun 2024; 92:e0037423. [PMID: 38289124 PMCID: PMC10929423 DOI: 10.1128/iai.00374-23] [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: 09/13/2023] [Accepted: 12/21/2023] [Indexed: 02/01/2024] Open
Abstract
Gamete surface protein P48/45 has been shown to be important for male gamete fertility and a strong candidate for the development of a malaria transmission-blocking vaccine (TBV). However, TBV development for Plasmodium vivax homolog Pvs48/45 has been slow because of a number of challenges: availability of conformationally suitable recombinant protein; the lack of an in vivo challenge model; and the inability to produce P. vivax gametocytes in culture to test transmission-blocking activity of antibodies. To support ongoing efforts to develop Pvs48/45 as a potential vaccine candidate, we initiated efforts to develop much needed reagents to move the field forward. We generated monoclonal antibodies (mAbs) directed against Pvs48/45 and characterized putative functional domains in Pvs48/45 using recombinant fragments corresponding to domains D1-D3 and their biological functionality through ex vivo direct membrane feeding assays (DMFAs) using P. vivax parasites from patients in a field setting in Brazil. While some mAbs partially blocked oocyst development in the DMFA, one mAb caused a significant enhancement of the infectivity of gametocytes in the mosquitoes. Individual mAbs exhibiting blocking and enhancing activities recognized non-overlapping epitopes in Pvs48/45. Further characterization of precise epitopes recognized by transmission-reducing and -enhancing antibodies will be crucial to design an effective immunogen with optimum transmission-reducing potential.
Collapse
Affiliation(s)
- Geetha P. Bansal
- Department of Tropical Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Maisa da Silva Araujo
- Plataforma de Produção e Infecção de Vetores da Malária, Laboratório de Entomologia - Fiocruz Rondônia, Porto Velho, Rondônia, Brazil
| | - Yi Cao
- Department of Tropical Medicine, Tulane University, New Orleans, Louisiana, USA
- Department of Global Health, George Washington University, Washington, DC, USA
| | - Emily Shaffer
- Department of Tropical Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Jessica Evangelista Araujo
- Plataforma de Produção e Infecção de Vetores da Malária, Laboratório de Entomologia - Fiocruz Rondônia, Porto Velho, Rondônia, Brazil
- Programa de Pós-Graduação em Biologia Experimental, Fundação Universidade Federal de Rondônia, Fiocruz Rondônia, Porto Velho, Rondônia, Brazil
| | - Jansen Fernandes Medeiros
- Plataforma de Produção e Infecção de Vetores da Malária, Laboratório de Entomologia - Fiocruz Rondônia, Porto Velho, Rondônia, Brazil
- Programa de Pós-Graduação em Biologia Experimental, Fundação Universidade Federal de Rondônia, Fiocruz Rondônia, Porto Velho, Rondônia, Brazil
| | - Clifford Hayashi
- Department of Global Health, George Washington University, Washington, DC, USA
| | - Joseph Vinetz
- Yale School of Medicine, New Haven, Connecticut, USA
| | - Nirbhay Kumar
- Department of Tropical Medicine, Tulane University, New Orleans, Louisiana, USA
- Department of Global Health, George Washington University, Washington, DC, USA
| |
Collapse
|
7
|
Kuesap J, Suphakhonchuwong N, Rungsihirunrat K. Genetic polymorphisms of Plasmodium vivax ookinete (sexual stage) surface proteins (Pvs25 and Pvs28) from Thailand. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2024; 118:105558. [PMID: 38244749 DOI: 10.1016/j.meegid.2024.105558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 01/16/2024] [Accepted: 01/17/2024] [Indexed: 01/22/2024]
Abstract
Plasmodium vivax is the most geographically widespread malaria parasite in human presently. The ookinete surface proteins of sexual stage of malaria parasites, Pvs25 and Pvs28, are candidates for the transmission blocking vaccine. The antigenic variation in population might be barrier for vaccine development. The objective of this study was to investigate the genetic diversity of Pvs25 and Pvs28 in endemic areas of Thailand. P. vivax clinical isolates collected from Thai-neighboring border areas were analyzed using polymerase chain reaction and sequencing method. Three and 14 amino acid substitutions were observed in 43 Pvs25 and 48 Pvs28 sequences, respectively. Three haplotypes in Pvs25 and 14 haplotypes with 5-7 GSGGE/D tandem repeats in Pvs28 were identified. The nucleotide diversity of pvs25 (π = 0.00059) had lower level than pvs28 (π = 0.00517). Tajima's D value for both pvs25 and pvs28 genes were negative while no significant difference was found (P > 0.10). Low genetic diversity was found in pvs25 and pvs28 genes in Thailand. The finding of the most frequent amino acid substitutions was consistent with global isolates. Therefore, the data could be helpful in developing of effective transmission blocking vaccine in malaria endemic areas.
Collapse
Affiliation(s)
- Jiraporn Kuesap
- Faculty of Allied Health Sciences, Thammasat University, Pathumthani, Thailand.
| | | | | |
Collapse
|
8
|
Bansal GP, Kumar N. Immune mechanisms targeting malaria transmission: opportunities for vaccine development. Expert Rev Vaccines 2024; 23:645-654. [PMID: 38888098 PMCID: PMC11472754 DOI: 10.1080/14760584.2024.2369583] [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: 02/06/2024] [Accepted: 06/14/2024] [Indexed: 06/20/2024]
Abstract
INTRODUCTION Malaria continues to remain a major global health problem with nearly a quarter of a billion clinical cases and more than 600,000 deaths in 2022. There has been significant progress toward vaccine development, however, poor efficacy of approved vaccines requiring multiple immunizing doses emphasizes the need for continued efforts toward improved vaccines. Progress to date, nonetheless, has provided impetus for malaria elimination. AREAS COVERED In this review we will focus on diverse immune mechanisms targeting gametocytes in the human host and gametocyte-mediated malaria transmission via the mosquito vector. EXPERT OPINION To march toward the goal of malaria elimination it will be critical to target the process of malaria transmission by mosquitoes, mediated exclusively by the sexual stages, i.e. male, and female gametocytes, ingested from infected vertebrate host. Studies over several decades have established antigens in the parasite sexual stages developing in the mosquito midgut as attractive targets for the development of transmission blocking vaccines (TBVs). Immune clearance of gametocytes in the vertebrate host can synergize with TBVs and directly aid in maintaining effective transmission reducing immune potential.
Collapse
Affiliation(s)
- Geetha P. Bansal
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, 70122, USA
| | - Nirbhay Kumar
- Department of Global Health, The Milken Institute School of Public Health, George Washington University, Washington DC, 20052, USA
| |
Collapse
|
9
|
Rajneesh, Tiwari R, Singh VK, Kumar A, Gupta RP, Singh AK, Gautam V, Kumar R. Advancements and Challenges in Developing Malaria Vaccines: Targeting Multiple Stages of the Parasite Life Cycle. ACS Infect Dis 2023; 9:1795-1814. [PMID: 37708228 DOI: 10.1021/acsinfecdis.3c00332] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/16/2023]
Abstract
Malaria, caused by Plasmodium species, remains a major global health concern, causing millions of deaths annually. While the introduction of the RTS,S vaccine has shown promise, there is a pressing need for more effective vaccines due to the emergence of drug-resistant parasites and insecticide-resistant vectors. However, the complex life cycle and genetic diversity of the parasite, technical obstacles, limited funding, and the impact of the 2019 pandemic have hindered progress in malaria vaccine development. This review focuses on advancements in malaria vaccine development, particularly the ongoing clinical trials targeting antigens from different stages of the Plasmodium life cycle. Additionally, we discuss the rationale, strategies, and challenges associated with vaccine design, aiming to enhance the immune response and protective efficacy of vaccine candidates. A cost-effective and multistage vaccine could hold the key to controlling and eradicating malaria.
Collapse
Affiliation(s)
- Rajneesh
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Rahul Tiwari
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Vishal K Singh
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Awnish Kumar
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Rohit P Gupta
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
- Department of Applied Microbiology, Institute of Science, Banaras Hindu University, Varanasi 221005, India
| | - Akhilesh K Singh
- Faculty of Dental Science, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Vibhav Gautam
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Rajiv Kumar
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| |
Collapse
|
10
|
Guissou E, Da DF, Hien DFDS, Yameogo KB, Yerbanga SR, Ouédraogo GA, Dabiré KR, Lefèvre T, Cohuet A. Intervention reducing malaria parasite load in vector mosquitoes: No impact on Plasmodium falciparum extrinsic incubation period and the survival of Anopheles gambiae. PLoS Pathog 2023; 19:e1011084. [PMID: 37195964 PMCID: PMC10191285 DOI: 10.1371/journal.ppat.1011084] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Accepted: 04/18/2023] [Indexed: 05/19/2023] Open
Abstract
In the fight against malaria, transmission blocking interventions (TBIs) such as transmission blocking vaccines or drugs, are promising approaches to complement conventional tools. They aim to prevent the infection of vectors and thereby reduce the subsequent exposure of a human population to infectious mosquitoes. The effectiveness of these approaches has been shown to depend on the initial intensity of infection in mosquitoes, often measured as the mean number of oocysts resulting from an infectious blood meal in absence of intervention. In mosquitoes exposed to a high intensity of infection, current TBI candidates are expected to be ineffective at completely blocking infection but will decrease parasite load and therefore, potentially also affect key parameters of vector transmission. The present study investigated the consequences of changes in oocyst intensity on subsequent parasite development and mosquito survival. To address this, we experimentally produced different intensities of infection for Anopheles gambiae females from Burkina Faso by diluting gametocytes from three natural Plasmodium falciparum local isolates and used a newly developed non-destructive method based on the exploitation of mosquito sugar feeding to track parasite and mosquito life history traits throughout sporogonic development. Our results indicate the extrinsic incubation period (EIP) of P. falciparum and mosquito survival did not vary with parasite density but differed significantly between parasite isolates with estimated EIP50 of 16 (95% CI: 15-18), 14 (95% CI: 12-16) and 12 (95% CI: 12-13) days and median longevity of 25 (95% CI: 22-29), 15 (95% CI: 13-15) and 18 (95% CI: 17-19) days for the three isolates respectively. Our results here do not identify unintended consequences of the decrease of parasite loads in mosquitoes on the parasite incubation period or on mosquito survival, two key parameters of vectorial capacity, and hence support the use of transmission blocking strategies to control malaria.
Collapse
Affiliation(s)
- Edwige Guissou
- Institut de Recherche en Sciences de la Santé, Bobo-Dioulasso, Burkina Faso
- MIVEGEC, Montpellier University, IRD, CNRS, Montpellier, France
- Université Nazi Boni, Bobo-Dioulasso, Burkina Faso
- Ecole Normale Supérieure, Koudougou, Burkina Faso
| | - Dari Frédéric Da
- Institut de Recherche en Sciences de la Santé, Bobo-Dioulasso, Burkina Faso
| | | | | | | | | | | | - Thierry Lefèvre
- Institut de Recherche en Sciences de la Santé, Bobo-Dioulasso, Burkina Faso
- MIVEGEC, Montpellier University, IRD, CNRS, Montpellier, France
| | - Anna Cohuet
- MIVEGEC, Montpellier University, IRD, CNRS, Montpellier, France
| |
Collapse
|
11
|
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
|
12
|
Wang S, Tian P, Li S, Liu H, Guo X, Huang F. Genetic diversity of transmission-blocking vaccine candidate antigens Pvs25 and Pvs28 in Plasmodium vivax isolates from China. BMC Infect Dis 2022; 22:944. [PMID: 36527077 PMCID: PMC9755777 DOI: 10.1186/s12879-022-07931-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Transmission-blocking vaccines (TBVs) target the sexual stages of malaria parasites to reduce or interrupt the transmission cycle in human and mosquito populations. The genetic diversity of TBVs candidate antigens, Pvs25 and Pvs28, in Plasmodium vivax could provide evidence for the development of TBVs. METHODS Dry blood spots from P. vivax patients were collected from Dandong, Suining, Hainan, Nyingchi, Tengchong, and Yingjiang in China. The pvs25 and pvs28 genes were amplified and sequenced. The genetic diversity of pvs25 and pvs28 were analyzed using DNASTAR, MEGA6, and DnaSP 5.0 programs. RESULTS A total of 377 samples were collected, among which 324 and 272 samples were successfully amplified in the pvs25 and pvs28 genes, respectively. Eight haplotypes were identified in Pvs25, for which the predominant mutation was I130T with 100% prevalence. A variety of 22 haplotypes in Pvs28 were identified. The number of GSGGE/D repeats of Pvs28 was a range of 4-8, among which, high (7-8) and low (4-5) copy numbers of tandem repeats were found in haplotypes H2 and H17, respectively. The nucleotide diversity of pvs28 (π = 0.00305 ± 0.00061) was slightly higher than that of pvs25 (π = 0.00146 ± 0.00007), thus they were not significantly different (P > 0.05). The Tajima's D value of pvs25 was positive whereas pvs28 was negative, which indicated that both genes were affected by natural selection. CONCLUSION The genetic diversity of pvs25 and pvs28 genes in China was relatively limited, which provided valuable information for TBVs design and optimization.
Collapse
Affiliation(s)
- Siqi Wang
- grid.198530.60000 0000 8803 2373National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, 200025 China ,grid.508378.1Chinese Center for Tropical Diseases Research, Shanghai, 200025 China ,grid.508378.1NHC Key Laboratory of Parasite and Vector Biology, National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, 200025 China ,grid.508378.1WHO Collaborating Centre for Tropical Diseases, National Center for International Research on Tropical Diseases, Shanghai, 200025 China
| | - Peng Tian
- grid.464500.30000 0004 1758 1139Yunnan Institute of Parasitic Diseases, Pu’er, 665000 China
| | - Shigang Li
- Yingjiang County Center for Disease Control and Prevention, Yingjiang, 679300 China
| | - Hui Liu
- grid.464500.30000 0004 1758 1139Yunnan Institute of Parasitic Diseases, Pu’er, 665000 China
| | - Xiangrui Guo
- Yingjiang County Center for Disease Control and Prevention, Yingjiang, 679300 China
| | - Fang Huang
- grid.430328.eShanghai Municipal Center for Disease Control and Prevention, Shanghai, 200336 China
| |
Collapse
|
13
|
Mulamba C, Williams C, Kreppel K, Ouedraogo JB, Olotu AI. Evaluation of the Pfs25-IMX313/Matrix-M malaria transmission-blocking candidate vaccine in endemic settings. Malar J 2022; 21:159. [PMID: 35655174 PMCID: PMC9161629 DOI: 10.1186/s12936-022-04173-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 05/02/2022] [Indexed: 11/10/2022] Open
Abstract
Malaria control relies heavily on the use of anti-malarial drugs and insecticides against malaria parasites and mosquito vectors. Drug and insecticide resistance threatens the effectiveness of conventional malarial interventions; alternative control approaches are, therefore, needed. The development of malaria transmission-blocking vaccines that target the sexual stages in humans or mosquito vectors is among new approaches being pursued. Here, the immunological mechanisms underlying malaria transmission blocking, status of Pfs25-based vaccines are viewed, as well as approaches and capacity for first in-human evaluation of a transmission-blocking candidate vaccine Pfs25-IMX313/Matrix-M administered to semi-immune healthy individuals in endemic settings. It is concluded that institutions in low and middle income settings should be supported to conduct first-in human vaccine trials in order to stimulate innovative research and reduce the overdependence on developed countries for research and local interventions against many diseases of public health importance.
Collapse
Affiliation(s)
- Charles Mulamba
- Interventions & Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania.,Nelson Mandela African Institution of Science and Technology, Tengeru, P. O. Box 447, Arusha, Tanzania
| | - Chris Williams
- The Jenner Institute, University of Oxford, Roosevelt Drive, Headington, Oxford, OX3 7DQ, UK
| | - Katharina Kreppel
- Nelson Mandela African Institution of Science and Technology, Tengeru, P. O. Box 447, Arusha, Tanzania
| | | | - Ally I Olotu
- Interventions & Clinical Trials Department, Ifakara Health Institute, P.O. Box 74, Bagamoyo, Tanzania.
| |
Collapse
|
14
|
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
|
15
|
Elucidating functional epitopes within the N-terminal region of malaria transmission blocking vaccine antigen Pfs230. NPJ Vaccines 2022; 7:4. [PMID: 35027567 PMCID: PMC8758780 DOI: 10.1038/s41541-021-00423-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 11/30/2021] [Indexed: 11/24/2022] Open
Abstract
Pfs230 is a leading malaria transmission blocking vaccine (TBV) candidate. Comprising 3135 amino acids (aa), the large size of Pfs230 necessitates the use of sub-fragments as vaccine immunogens. Therefore, determination of which regions induce functional antibody responses is essential. We previously reported that of 27 sub-fragments spanning the entire molecule, only five induced functional antibodies. A “functional” antibody is defined herein as one that inhibits Plasmodium falciparum parasite development in mosquitoes in a standard membrane-feeding assay (SMFA). These five sub-fragments were found within the aa 443–1274 range, and all contained aa 543–730. Here, we further pinpoint the location of epitopes within Pfs230 that are recognized by functional antibodies using antibody depletion and enrichment techniques. Functional epitopes were not found within the aa 918–1274 region. Within aa 443–917, further analysis showed the existence of functional epitopes not only within the aa 543–730 region but also outside of it. Affinity-purified antibodies using a synthetic peptide matching aa 543–588 showed activity in the SMFA. Immunization with a synthetic peptide comprising this segment, formulated either as a carrier-protein conjugate vaccine or with a liposomal vaccine adjuvant system, induced antibodies in mice that were functional in the SMFA. These findings provide key insights for Pfs230-based vaccine design and establish the feasibility for the use of synthetic peptide antigens for a malaria TBV.
Collapse
|
16
|
Arévalo-Herrera M, Miura K, Solano E, Ramírez JS, Long CA, Corradin G, Herrera S. Immunogenicity of full-length P. vivax rPvs48/45 protein formulations in BALB/c mice. Vaccine 2022; 40:133-140. [PMID: 34802791 PMCID: PMC9109962 DOI: 10.1016/j.vaccine.2021.11.036] [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] [Received: 06/10/2021] [Revised: 10/12/2021] [Accepted: 11/11/2021] [Indexed: 01/05/2023]
Abstract
BACKGROUND Pvs48/45 is a Plasmodium vivax gametocyte surface protein involved in the parasite fertilization process. Previous studies showed that Pvs48/45 proteins expressed in Escherichia coli (E. coli) and Chinese hamster ovary (CHO) cells were highly immunoreactive with sera from malaria-endemic areas and highly immunogenic in animal models. Here the immunogenicity in mice of three different vaccine formulations was compared. METHODS Recombinant (r) Pvs48/45 proteins were expressed in E. coli and CHO, purified, formulated in Alhydrogel, GLA-SE and Montanide ISA-51 adjuvants and used to immunize BALB/c mice. Animals were immunized on days 0, 20 and 40, and serum samples were collected for serological analyses of specific antibody responses using ELISA and immunofluorescence (IFAT). Additionally, ex-vivo transmission-reducing activity (TRA) of sera on P. vivax gametocyte-infected human blood fed to Anopheles albimanus in direct membrane feeding assays (DMFA) was evaluated. RESULTS Most immunized animals seroconverted after the first immunization, and some developed antibody peaks of 106 with all adjuvants. However, the three adjuvant formulations induced different antibody responses and TRA efficacy. While GLA-SE formulations of both proteins induced similar antibody profiles, Montanide ISA-51 formulations resulted in higher and longer-lasting antibody titers with CHO-rPvs48/45 than with the E. coli formulation. Although the CHO protein formulated in Alhydrogel generated a high initial antibody peak, antibody responses to both proteins rapidly waned. Likewise, anti-Pvs48/45 antibodies displayed differential recognition of the parasite proteins in IFAT and ex vivo blockade of parasite transmission to mosquitoes. The CHO-rPvs48/45 formulated in Montanide ISA-51 induced the most effective ex vivo parasite blockage. CONCLUSIONS Three out of six vaccine formulations elicited antibodies with ex vivo TRA. The CHO-rPvs48/45 Montanide ISA-51 formulation induced the most stable antibody response, recognizing the native protein and the most robust ex vivo TRA. These results encourage further testing of the vaccine potential of this protein.
Collapse
Affiliation(s)
- Myriam Arévalo-Herrera
- Malaria Vaccine and Drug Development Center, Cali, Colombia.,Caucaseco Scientific Research Center, Cali, Colombia.,Corresponding author: Myriam Arevalo-Herrera, PhD, , Phone: (+57 2) 521 6232, Fax: (+57 2) 521 6228, Malaria Vaccine and Drug Development Center, Carrera 37 2 Bis # 5E-08. Cali, Colombia
| | - 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
| | | | - Sócrates Herrera
- Malaria Vaccine and Drug Development Center, Cali, Colombia.,Caucaseco Scientific Research Center, Cali, Colombia
| |
Collapse
|
17
|
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
|
18
|
Memariani H, Memariani M. Melittin as a promising anti-protozoan peptide: current knowledge and future prospects. AMB Express 2021; 11:69. [PMID: 33983454 PMCID: PMC8119515 DOI: 10.1186/s13568-021-01229-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 05/05/2021] [Indexed: 12/19/2022] Open
Abstract
Protozoan diseases such as malaria, leishmaniasis, Chagas disease, and sleeping sickness still levy a heavy toll on human lives. Deplorably, only few classes of anti-protozoan drugs have thus far been developed. The problem is further compounded by their intrinsic toxicity, emergence of drug resistance, and the lack of licensed vaccines. Thus, there is a genuine exigency to develop novel anti-protozoan medications. Over the past years, melittin, the major constituent in the venom of European honeybee Apis mellifera, has gathered the attention of researchers due to its potential therapeutic applications. Insofar as we are aware, there has been no review pertinent to anti-protozoan properties of melittin. The present review outlines the current knowledge about anti-protozoan effects of melittin and its underlying mechanisms. The peptide has proven to be efficacious in killing different protozoan parasites such as Leishmania, Plasmodium, Toxoplasma, and Trypanosoma in vitro. Apart from direct membrane-disruptive activity, melittin is capable of destabilizing calcium homeostasis, reducing mitochondrial membrane potential, disorganizing kinetoplast DNA, instigating apoptotic cell death, and induction of autophagy in protozoan pathogens. Emerging evidence suggests that melittin is a promising candidate for future vaccine adjuvants. Transmission-blocking activity of melittin against vector-borne pathogens underscores its potential utility for both transgenic and paratransgenic manipulations. Nevertheless, future research should focus upon investigating anti-microbial activities of melittin, alone or in combination with the current anti-protozoan medications, against a far broader spectrum of protozoan parasites as well as pre-clinical testing of the peptide in animal models.
Collapse
|
19
|
Pirahmadi S, Zakeri S, Djadid ND, Mehrizi AA. A review of combination adjuvants for malaria vaccines: a promising approach for vaccine development. Int J Parasitol 2021; 51:699-717. [PMID: 33798560 DOI: 10.1016/j.ijpara.2021.01.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 12/18/2020] [Accepted: 01/28/2021] [Indexed: 01/16/2023]
Abstract
It is obvious that there is a critical need for an efficient malaria vaccine to accelerate malaria eradication. Currently, recombinant subunit vaccination against malaria using proteins and peptides is gaining attention. However, one of the major drawbacks of this approach is the lack of an efficient and durable immune response. Therefore, subunit vaccines require adjuvants to make the vaccine sufficiently immunogenic. Considering the history of the RTS,S vaccine, it seems likely that no single adjuvant is capable of eliciting all the protective immune responses required in many malarial subunit vaccines and the use of combination adjuvants will be increasingly important as the science of malaria vaccines advances. In light of this, it appears that identifying the most effective mixture of adjuvants with minimal adverse effects offers tremendous opportunities in improving the efficacy of vaccines against malaria. Owing to the importance of a multi-adjuvanted approach in subunit malaria vaccine development, this review paper outlines some of the best known combination adjuvants used in malaria subunit vaccines, focusing on their proposed mechanisms of action, their immunological properties, and their notable results. The aim of the present review is to consolidate these findings to aid the application of these combination adjuvants in experimental malaria vaccines.
Collapse
Affiliation(s)
- Sakineh Pirahmadi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Sedigheh Zakeri
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran.
| | - Navid D Djadid
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Akram A Mehrizi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
20
|
Lozano JM, Rodríguez Parra Z, Hernández-Martínez S, Yasnot-Acosta MF, Rojas AP, Marín-Waldo LS, Rincón JE. The Search of a Malaria Vaccine: The Time for Modified Immuno-Potentiating Probes. Vaccines (Basel) 2021; 9:vaccines9020115. [PMID: 33540947 PMCID: PMC7913233 DOI: 10.3390/vaccines9020115] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/22/2021] [Accepted: 01/25/2021] [Indexed: 12/25/2022] Open
Abstract
Malaria is a deadly disease that takes the lives of more than 420,000 people a year and is responsible for more than 229 million clinical cases globally. In 2019, 95% of malaria morbidity occurred in African countries. The development of a highly protective vaccine is an urgent task that remains to be solved. Many vaccine candidates have been developed, from the use of the entire attenuated and irradiated pre-erythrocytic parasite forms (or recombinantly expressed antigens thereof) to synthetic candidates formulated in a variety of adjuvants and delivery systems, however these have unfortunately proven a limited efficacy. At present, some vaccine candidates are finishing safety and protective efficacy trials, such as the PfSPZ and the RTS,S/AS01 which are being introduced in Africa. We propose a strategy for introducing non-natural elements into target antigens representing key epitopes of Plasmodium spp. Accordingly, chemical strategies and knowledge of host immunity to Plasmodium spp. have served as the basis. Evidence is obtained after being tested in experimental rodent models for malaria infection and recognized for human sera from malaria-endemic regions. This encourages us to propose such an immune-potentiating strategy to be further considered in the search for new vaccine candidates.
Collapse
Affiliation(s)
- José Manuel Lozano
- Grupo de Investigación Mimetismo Molecular de los Agentes Infecciosos, Departamento de Farmacia, Universidad Nacional de Colombia—Sede Bogotá, 111321 Bogota, Colombia;
- Correspondence: ; Tel.: +57-3102-504-657
| | - Zully Rodríguez Parra
- Grupo de Investigación Mimetismo Molecular de los Agentes Infecciosos, Departamento de Farmacia, Universidad Nacional de Colombia—Sede Bogotá, 111321 Bogota, Colombia;
| | - Salvador Hernández-Martínez
- Dirección de Infección e Inmunidad, Centro de Investigaciones Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, 62508 Cuernavaca, Morelos, Mexico;
| | - Maria Fernanda Yasnot-Acosta
- Grupo de Investigaciones Microbiológicas y Biomédicas de Córdoba, Universidad de Córdoba, 230002 Monteria, Colombia;
| | - Angela Patricia Rojas
- Grupo de Investigación Biología Celular y Autoinmuniad, Departamento de Farmacia, Universidad Nacional de Colombia-Sede Bogotá, 111321 Bogota, Colombia;
| | | | - Juan Edilberto Rincón
- Departamento de Ingeniería y Mecatrónica, Universidad Nacional de Colombia-Sede Bogotá, 111321 Bogota, Colombia;
| |
Collapse
|
21
|
Liu F, Yang F, Wang Y, Hong M, Zheng W, Min H, Li D, Jin Y, Tsuboi T, Cui L, Cao Y. A conserved malaria parasite antigen Pb22 plays a critical role in male gametogenesis in Plasmodium berghei. Cell Microbiol 2020; 23:e13294. [PMID: 33222390 DOI: 10.1111/cmi.13294] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 10/28/2020] [Accepted: 11/16/2020] [Indexed: 12/13/2022]
Abstract
Gametogenesis, the formation of gametes from gametocytes, an essential step for malaria parasite transmission, is targeted by transmission-blocking drugs and vaccines. We identified a conserved protein (PBANKA_0305900) in Plasmodium berghei, which encodes a protein of 22 kDa (thus named Pb22) and is expressed in both asexual stages and gametocytes. Its homologues are present in all Plasmodium species and its closely related, Hepatocystis, but not in other apicomplexans. Pb22 protein was localised in the cytosols of schizonts, as well as male and female gametocytes. During gamete-to-ookinete development, Pb22 became localised on the plasma membranes of gametes and ookinetes. Compared to the wild-type (WT) parasites, P. berghei with pb22 knockout (KO) showed a significant reduction in exflagellation (~89%) of male gametocytes and ookinete number (~97%) during in vitro ookinete culture. Mosquito feeding assays showed that ookinete and oocyst formation of the pb22-KO line in mosquito midguts was almost completely abolished. These defects were rescued in parasites where pb22 was restored. Cross-fertilisation experiments with parasite lines defective in either male or female gametes confirmed that the defects in the pb22-KO line were restricted to the male gametes, whereas female gametes in the pb22-KO line were fertile at the WT level. Detailed analysis of male gametogenesis showed that 30% of the male gametocytes in the pb22-KO line failed to assemble the axonemes, whereas ~48.9% of the male gametocytes formed flagella but failed to egress from the host erythrocyte. To explore its transmission-blocking potential, recombinant Pb22 (rPb22) was expressed and used to immunise mice. in vitro assays showed that the rPb22-antisera significantly inhibited exflagellation by ~64.8% and ookinete formation by ~93.4%. Mosquitoes after feeding on rPb22-immunised mice also showed significant decreases in infection prevalence (83.3-93.3%) and oocyst density (93.5-99.6%). Further studies of the Pb22 orthologues in human malaria parasites are warranted.
Collapse
Affiliation(s)
- Fei Liu
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Fan Yang
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Yaru Wang
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Minsheng Hong
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Wenqi Zheng
- Department of Clinical Laboratory, Affiliated Hospital of Inner Mongolian Medical University, Hohhot, China
| | - Hui Min
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China.,Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Danni Li
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Ying Jin
- Division of Administration, Liaoning Research Institute of Family Planning, Shenyang, China
| | - Takafumi Tsuboi
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - 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
| |
Collapse
|
22
|
Scaria PV, Chen BB, Rowe CG, Alani N, Muratova OV, Barnafo EK, Lambert LE, Zaidi IU, Lees A, Rausch KM, Narum DL, Duffy PE. Comparison of carrier proteins to conjugate malaria transmission blocking vaccine antigens, Pfs25 and Pfs230. Vaccine 2020; 38:5480-5489. [PMID: 32600913 PMCID: PMC11127250 DOI: 10.1016/j.vaccine.2020.06.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 06/03/2020] [Accepted: 06/05/2020] [Indexed: 12/18/2022]
Abstract
Malaria transmission blocking vaccines (TBV) target the sexual stage of the parasite and have been pursued as a stand-alone vaccine or for combination with pre-erythrocytic or blood stage vaccines. Our efforts to develop TBV focus primarily on two antigens, Pfs25 and Pfs230. Chemical conjugation of these poorly immunogenic antigens to carrier proteins enhances their immunogenicity, and conjugates of these antigens to Exoprotein A (EPA) are currently under evaluation in clinical trials. Nonetheless, more potent carriers may augment the immunogenicity of these antigens for a more efficacious vaccine; here, we evaluate a series of proteins to identify such a carrier. Pfs25 and Pfs230 were chemically conjugated to 4 different carriers [tetanus toxoid (TT), a recombinant fragment of tetanus toxin heavy chain (rTThc), recombinant CRM197 produced in Pseudomonas fluorescens (CRM197) or in E. coli (EcoCRM®)] and compared to EPA conjugates in mouse immunogenicity studies. Conjugates of each antigen formulated in Alhydrogel® elicited similar antibody titers but showed differences in functional activity. At a 0.5 µg dose, Pfs230 conjugated to TT, CRM197 and EcoCRM® showed significantly higher functional activity compared to EPA. When formulated with the more potent adjuvant GLA-LSQ, all 4 alternate conjugates induced higher antibody titers as well as increased functional activity compared to the EPA conjugate. IgG subclass analysis of Pfs230 conjugates showed no carrier-dependent differences in the IgG profile. While Alhydrogel® formulations induced a Th2 dominant immune response, GLA-LSQ formulations induced a mixed Th1/Th2 response.
Collapse
Affiliation(s)
- Puthupparampil V Scaria
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Beth B Chen
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Christopher G Rowe
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Nada Alani
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Olga V Muratova
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Emma K Barnafo
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Lynn E Lambert
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Irfan U Zaidi
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | - Kelly M Rausch
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - David L Narum
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Patrick E Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
23
|
Zheng W, Liu F, Du F, Yang F, Kou X, He Y, Feng H, Fan Q, Luo E, Min H, Miao J, Cui L, Cao Y. Characterization of a Sulfhydryl Oxidase From Plasmodium berghei as a Target for Blocking Parasite Transmission. Front Cell Infect Microbiol 2020; 10:311. [PMID: 32670896 PMCID: PMC7332561 DOI: 10.3389/fcimb.2020.00311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 05/22/2020] [Indexed: 11/13/2022] Open
Abstract
Quiescin sulfhydryl oxidase (QSOX), present in a wide variety of eukaryotic species, catalyzes the insertion of disulfide bonds into unfolded, reduced proteins. Here we characterized the QSOX protein from the rodent malaria parasite Plasmodium berghei (PbQSOX), which is conserved in all sequenced malaria parasite species. The PbQSOX protein was not expressed in asexual erythrocytic stages, but was most abundantly expressed in ookinetes. Indirect immunofluorescence assays revealed PbQSOX was not only localized in cytoplasm of gametocytes, gametes and ookinetes, but also expressed on the surface of gametes and ookinetes. Western blot identified extracellular presence of PbQSOX in the culture medium of ookinetes suggestive of secretion. Pbqsox deletion (Δpbqsox) did not affect asexual intraerythrocytic development, but reduced exflagellation of male gametocytes as well as formation and maturation of ookinetes. Pbqsox deletion also led to a significant increase in the reduced thiol groups of ookinete surface proteins, suggesting that it may play a role in maintaining the integrity of disulfide bonds of surface proteins, which might be needed for ookinete development. Mosquitoes that fed on Δpbqsox-infected mice showed a significant reduction in ookinete and oocyst numbers compared to those fed on wild-type parasite-infected mice. Further, both polyclonal mouse antisera and a monoclonal antibody against the recombinant PbQSOX exhibited substantial transmission-blocking activities in in vitro and mosquito feeding assays, suggesting QSOX is a potential target for blocking parasite transmission.
Collapse
Affiliation(s)
- Wenqi Zheng
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
- Department of Clinical Laboratory, Affiliated Hospital of Inner Mongolian Medical University, Hohhot, China
| | - Fei Liu
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Feng Du
- Department of Pathogen Biology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Fan Yang
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Xu Kou
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
- Department of Animal Quarantine, College of Animal Husbandry and Veterinary Sciences, Liaoning Medical University, Jinzhou, China
| | - Yiwen He
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Hui Feng
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Qi Fan
- Dalian Institute of Biotechnology, Dalian, China
| | - Enjie Luo
- Department of Pathogen Biology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Hui Min
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Jun Miao
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Liwang Cui
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Yaming Cao
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| |
Collapse
|
24
|
Choi R, Michaels SA, Onu EC, Hulverson MA, Saha A, Coker ME, Weeks JC, Van Voorhis WC, Ojo KK. Taming the Boys for Global Good: Contraceptive Strategy to Stop Malaria Transmission. Molecules 2020; 25:molecules25122773. [PMID: 32560085 PMCID: PMC7356879 DOI: 10.3390/molecules25122773] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/10/2020] [Accepted: 06/14/2020] [Indexed: 12/21/2022] Open
Abstract
Transmission of human malaria parasites (Plasmodium spp.) by Anopheles mosquitoes is a continuous process that presents a formidable challenge for effective control of the disease. Infectious gametocytes continue to circulate in humans for up to four weeks after antimalarial drug treatment, permitting prolonged transmission to mosquitoes even after clinical cure. Almost all reported malaria cases are transmitted to humans by mosquitoes, and therefore decreasing the rate of Plasmodium transmission from humans to mosquitoes with novel transmission-blocking remedies would be an important complement to other interventions in reducing malaria incidence.
Collapse
Affiliation(s)
- Ryan Choi
- Center for Emerging and Re-Emerging Infectious Diseases (CERID), Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA 98109, USA; (R.C.); (S.A.M.); (M.A.H.); (A.S.); (W.C.V.V.)
| | - Samantha A. Michaels
- Center for Emerging and Re-Emerging Infectious Diseases (CERID), Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA 98109, USA; (R.C.); (S.A.M.); (M.A.H.); (A.S.); (W.C.V.V.)
| | - Emmanuel C. Onu
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, University of Ibadan, Ibadan, Nigeria; (E.C.O.); (M.E.C.)
| | - Matthew A. Hulverson
- Center for Emerging and Re-Emerging Infectious Diseases (CERID), Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA 98109, USA; (R.C.); (S.A.M.); (M.A.H.); (A.S.); (W.C.V.V.)
| | - Aparajita Saha
- Center for Emerging and Re-Emerging Infectious Diseases (CERID), Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA 98109, USA; (R.C.); (S.A.M.); (M.A.H.); (A.S.); (W.C.V.V.)
| | - Morenike E. Coker
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, University of Ibadan, Ibadan, Nigeria; (E.C.O.); (M.E.C.)
| | - Janis C. Weeks
- Department of Biology, University of Oregon, Eugene, OR 97403, USA;
| | - Wesley C. Van Voorhis
- Center for Emerging and Re-Emerging Infectious Diseases (CERID), Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA 98109, USA; (R.C.); (S.A.M.); (M.A.H.); (A.S.); (W.C.V.V.)
| | - Kayode K. Ojo
- Center for Emerging and Re-Emerging Infectious Diseases (CERID), Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA 98109, USA; (R.C.); (S.A.M.); (M.A.H.); (A.S.); (W.C.V.V.)
- Correspondence: ; Tel.: +1-206-543-0821
| |
Collapse
|
25
|
Li D, Yu C, Guo J, Wang Y, Zhao Y, Wang L, Soe MT, Feng H, Kyaw MP, Sattabongkot J, Jiang L, Cui L, Zhu X, Cao Y. Plasmodium vivax HAP2/GCS1 gene exhibits limited genetic diversity among parasite isolates from the Greater Mekong Subregion. Parasit Vectors 2020; 13:175. [PMID: 32264948 PMCID: PMC7137254 DOI: 10.1186/s13071-020-04050-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 03/28/2020] [Indexed: 12/02/2022] Open
Abstract
Background Antigens expressed in sexual stages of the malaria parasites are targets of transmission-blocking vaccines (TBVs). HAP2/GCS1, a TBV candidate, is critical for fertilization in Plasmodium. Here, the genetic diversity of PvHAP2 was studied in Plasmodium vivax parasite populations from the Greater Mekong Subregion (GMS). Methods Plasmodium vivax clinical isolates were collected in clinics from the China-Myanmar border region (135 samples), western Thailand (41 samples) and western Myanmar (51 samples). Near full-length Pvhap2 (nucleotides 13–2574) was amplified and sequenced from these isolates. Molecular evolution studies were conducted to evaluate the genetic diversity, selection and population differentiation. Results Sequencing of the pvhap2 gene for a total of 227 samples from the three P. vivax populations revealed limited genetic diversity of this gene in the GMS (π = 0.00036 ± 0.00003), with the highest π value observed in Myanmar (0.00053 ± 0.00009). Y133S was the dominant mutation in the China-Myanmar border (99.26%), Myanmar (100%) and Thailand (95.12%). Results of all neutrality tests were negative for all the three populations, suggesting the possible action of purifying selection. Codon-based tests identified specific codons which are under purifying or positive selections. Wright’s fixation index showed low to moderate genetic differentiation of P. vivax populations in the GMS, with FST ranging from 0.04077 to 0.24833, whereas high levels of genetic differentiation were detected between the China-Myanmar border and Iran populations (FST = 0.60266), and between Thailand and Iran populations (FST = 0.44161). A total of 20 haplotypes were identified, with H2 being the abundant haplotype in China-Myanmar border, Myanmar and Thailand populations. Epitope mapping prediction of Pvhap2 antigen showed that high-score B-cell epitopes are located in the S307-G324, L429-P453 and V623-D637 regions. The E317K and D637N mutations located within S307-G324 and V623-D637 epitopes slightly reduced the predicted score for potential epitopes. Conclusions The present study showed a very low level of genetic diversity of pvhap2 gene among P. vivax populations in the Greater Mekong Subregion. The relative conservation of pvhap2 supports further evaluation of a Pvhap2-based TBV.![]()
Collapse
Affiliation(s)
- Danni Li
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, 110122, Liaoning Province, People's Republic of China
| | - Chunyun Yu
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, 110122, Liaoning Province, People's Republic of China
| | - Jian Guo
- Department of Laboratory Medicine, Shanghai East Hospital, Tongji School of Medicine, Shanghai, People's Republic of China
| | - Yazhou Wang
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, 110122, Liaoning Province, People's Republic of China
| | - Yan Zhao
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, 110122, Liaoning Province, People's Republic of China
| | - Lin Wang
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, 110122, Liaoning Province, People's Republic of China
| | - Myat Thu Soe
- Myanmar Health Network Organization, Yangon, Myanmar
| | - Hui Feng
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, 110122, Liaoning Province, People's Republic of China
| | | | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Lubin Jiang
- Unit of Human Parasite Molecular and Cell Biology, Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Liwang Cui
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, 3720 Spectrum Boulevard, Suite 304, Tampa, FL, 33612, USA
| | - Xiaotong Zhu
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, 110122, Liaoning Province, People's Republic of China.
| | - Yaming Cao
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, 110122, Liaoning Province, People's Republic of China.
| |
Collapse
|
26
|
Qiu Y, Zhao Y, Liu F, Ye B, Zhao Z, Thongpoon S, Roobsoong W, Sattabongkot J, Cui L, Fan Q, Cao Y. Evaluation of Plasmodium vivax HAP2 as a transmission-blocking vaccine candidate. Vaccine 2020; 38:2841-2848. [PMID: 32093983 DOI: 10.1016/j.vaccine.2020.02.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 01/31/2020] [Accepted: 02/05/2020] [Indexed: 12/23/2022]
Abstract
Transmission-blocking vaccine (TBV) is a promising strategy to interfere with the transmission of malaria. To date, only limited TBV candidate antigens have been identified for Plasmodium vivax. HAP2 is a gamete membrane fusion protein, with homology to the class II viral fusion proteins. Herein we reported the characterization of the PvHAP2 for its potential as a TBV candidate for P. vivax. The HAP2/GCS1 domain of PvHAP2 was expressed in the baculovirus expression system and the recombinant protein was used to raise antibodies in rabbits. Indirect immunofluorescence assays showed that anti-PvHAP2 antibodies reacted only with the male gametocytes on blood smears. Direct membrane feeding assays were conducted using four field P. vivax isolates in Anopheles dirus. At a mean infection intensity of 72.4, 70.7, 51.3, and 15.6 oocysts/midgut with the control antibodies, anti-PvHAP2 antibodies significantly reduced the midgut oocyst intensity by 40.3, 44.4, 61.9, and 89.7%. Whereas the anti-PvHAP2 antibodies were not effective in reducing the infection prevalence at higher parasite exposure (51.3-72.4 oocysts/midgut in the control group), the anti-PvHAP2 antibodies reduced infection prevalence by 50% at a low challenge (15.6 oocysts/midgut). Multiple sequence alignment showed 100% identity among these Thai P. vivax isolates, suggesting that polymorphism may not be an impediment for the utilization of PvHAP2 as a TBV antigen. In conclusion, our results suggest that PvHAP2 could serve as a TBV candidate for P. vivax, and further optimization and evaluation are warranted.
Collapse
Affiliation(s)
- Yue Qiu
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, Liaoning 110122, China
| | - Yan Zhao
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, Liaoning 110122, China
| | - Fei Liu
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, Liaoning 110122, China
| | - Bo Ye
- Dalian Institute of Biotechnology, Dalian, Liaoning, China
| | - Zhenjun Zhao
- Dalian Institute of Biotechnology, Dalian, Liaoning, China
| | - Sataporn Thongpoon
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Thailand
| | - Wanlapa Roobsoong
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Thailand
| | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Thailand
| | - Liwang Cui
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, 3720 Spectrum Boulevard, Suite 304, Tampa, FL 33612, USA
| | - Qi Fan
- Dalian Institute of Biotechnology, Dalian, Liaoning, China
| | - Yaming Cao
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, Liaoning 110122, China.
| |
Collapse
|
27
|
Ararat-Sarria M, Prado CC, Camargo M, Ospina LT, Camargo PA, Curtidor H, Patarroyo MA. Sexual forms obtained in a continuous in vitro cultured Colombian strain of Plasmodium falciparum (FCB2). Malar J 2020; 19:57. [PMID: 32014000 PMCID: PMC6998264 DOI: 10.1186/s12936-020-3142-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 01/25/2020] [Indexed: 12/03/2022] Open
Abstract
Background The epidemiological control of malaria has been hampered by the appearance of parasite resistance to anti-malarial drugs and by the resistance of mosquito vectors to control measures. This has also been associated with weak transmission control, mostly due to poor control of asymptomatic patients associated with host-vector transmission. This highlights the importance of studying the parasite’s sexual forms (gametocytes) which are involved in this phase of the parasite’s life-cycle. Some African and Asian strains of Plasmodium falciparum have been fully characterized regarding sexual forms’ production; however, few Latin-American strains have been so characterized. This study was aimed at characterizing the Colombian FCB2 strain as a gametocyte producer able to infect mosquitoes. Methods Gametocyte production was induced in in vitro cultured P. falciparum FCB2 and 3D7 strains. Pfap2g and Pfs25 gene expression was detected in FCB2 strain gametocyte culture by RT-PCR. Comparative analysis of gametocytes obtained from both strains was made (counts and morphological changes). In vitro zygote formation from FCB2 gametocytes was induced by incubating a gametocyte culture sample at 27 °C for 20 min. A controlled Anopheles albimanus infection was made using an artificial feed system with cultured FCB2 gametocytes (14–15 days old). Mosquito midgut dissection was then carried out for analyzing oocysts. Results The FCB2 strain expressed Pfap2g, Pfs16, Pfg27/25 and Pfs25 sexual differentiation-related genes after in vitro sexual differentiation induction, producing gametocytes that conserved the expected morphological features. The amount of FCB2 gametocytes produced was similar to that from the 3D7 strain. FCB2 gametocytes were differentiated into zygotes and ookinetes after an in vitro low-temperature stimulus and infected An. albimanus mosquitoes, developing to oocyst stage. Conclusions Even with the history of long-term FCB2 strain in vitro culture maintenance, it has retained its sexual differentiation ability. The gametocytes produced here preserved these parasite forms’ usual characteristics and An. albimanus infection capability, thus enabling its use as a tool for studying sexual form biology, An. albimanus infection comparative analysis and anti-malarial drug and vaccine development.
Collapse
Affiliation(s)
- Monica Ararat-Sarria
- Receptor-Ligand Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia.,PhD Programme in Biomedical and Biological Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Cesar Camilo Prado
- Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia
| | - Milena Camargo
- Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia
| | - Laura Tatiana Ospina
- Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia
| | - Paola Andrea Camargo
- Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia
| | - Hernando Curtidor
- Receptor-Ligand Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia.,Animal Science Faculty, Universidad de Ciencias Aplicadas y Ambientales (U.D.C.A), Bogotá, Colombia
| | - Manuel Alfonso Patarroyo
- Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia. .,School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia.
| |
Collapse
|
28
|
Miura K, Tachibana M, Takashima E, Morita M, Kanoi BN, Nagaoka H, Baba M, Torii M, Ishino T, Tsuboi T. Malaria transmission-blocking vaccines: wheat germ cell-free technology can accelerate vaccine development. Expert Rev Vaccines 2019; 18:1017-1027. [PMID: 31566026 PMCID: PMC11000147 DOI: 10.1080/14760584.2019.1674145] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Accepted: 09/25/2019] [Indexed: 12/18/2022]
Abstract
Introduction: Highly effective malaria vaccines are essential component toward malaria elimination. Although the leading malaria vaccine, RTS,S/AS01, with modest efficacy is being evaluated in a pilot feasibility trial, development of a malaria transmission-blocking vaccine (TBV) could make a major contribution toward malaria elimination. Only a few TBV antigens have reached pre-clinical or clinical development but with several challenges including difficulties in the expression of malaria recombinant proteins and low immunogenicity in humans. Therefore, novel approaches to accelerate TBV research to preclinical development are critical to generate an efficacious TBV.Areas covered: PubMed was searched to review the progress and future prospects of malaria TBV research and development. We also reviewed registered trials at ClinicalTrials.gov as well as post-genome TBV candidate discovery research including our efforts.Expert opinion: Wheat germ cell-free protein synthesis technology can accelerate TBV development by overcoming some current challenges of TBV research.
Collapse
Affiliation(s)
- Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Mayumi Tachibana
- Division of Molecular Parasitology, Proteo-Science Center, Ehime University, Toon, Japan
| | - Eizo Takashima
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Masayuki Morita
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Bernard N Kanoi
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Hikaru Nagaoka
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Minami Baba
- Division of Molecular Parasitology, Proteo-Science Center, Ehime University, Toon, Japan
| | - Motomi Torii
- Division of Molecular Parasitology, Proteo-Science Center, Ehime University, Toon, Japan
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Tomoko Ishino
- Division of Molecular Parasitology, Proteo-Science Center, Ehime University, Toon, Japan
| | - Takafumi Tsuboi
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| |
Collapse
|
29
|
Liu F, Liu Q, Yu C, Zhao Y, Wu Y, Min H, Qiu Y, Jin Y, Miao J, Cui L, Cao Y. An MFS-Domain Protein Pb115 Plays a Critical Role in Gamete Fertilization of the Malaria Parasite Plasmodium berghei. Front Microbiol 2019; 10:2193. [PMID: 31616399 PMCID: PMC6764285 DOI: 10.3389/fmicb.2019.02193] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Accepted: 09/06/2019] [Indexed: 12/20/2022] Open
Abstract
Sexual reproduction is an essential process in the Plasmodium life cycle and a vulnerable step for blocking transmission from the human host to mosquitoes. In this study, we characterized the functions of a conserved cell membrane protein P115 in the rodent malaria parasite Plasmodium berghei ANKA. Pb115 was expressed in both asexual stages (schizonts) and sexual stages (gametocytes, gametes, and ookinetes), and was localized on the plasma membrane of gametes and ookinetes. In P. berghei, genetic deletion of Pb115 (Δpb115) did not affect asexual multiplication, nor did it affect gametocyte development or exflagellation of the male gametocytes. However, mosquitoes fed on Δpb115-infected mice showed 74% reduction in the prevalence of infection and 96.5% reduction in oocyst density compared to those fed on wild-type P. berghei-infected mice. The Δpb115 parasites showed significant defects in the interactions between the male and female gametes, and as a result, very few zygotes were formed in ookinete cultures. Cross fertilization with the male-defective Δpbs48/45 line and the female-defective Δpfs47 line further indicated that the fertilization defects of the Δpb115 lines were present in both male and female gametes. We evaluated the transmission-blocking potential of Pb115 by immunization of mice with a recombinant Pb115 fragment. In vivo mosquito feeding assay showed Pb115 immunization conferred modest, but significant transmission reducing activity with 44% reduction in infection prevalence and 39% reduction in oocyst density. Our results described functional characterization of a conserved membrane protein as a fertility factor in Plasmodium and demonstrated transmission-blocking potential of this antigen.
Collapse
Affiliation(s)
- Fei Liu
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Qingyang Liu
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Chunyun Yu
- 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
| | - Yudi Wu
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Hui Min
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Tampa, FL, United States
| | - Yue Qiu
- The First Hospital of China Medical University, Shenyang, China
| | - Ying Jin
- Liaoning Research Institute of Family Planning, Shenyang, China
| | - Jun Miao
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Tampa, FL, United States
| | - Liwang Cui
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Tampa, FL, United States
| | - Yaming Cao
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| |
Collapse
|
30
|
Jakhar R, Kumar P, Sehrawat N, Gakhar SK. A comprehensive analysis of amino-peptidase N1 protein (APN) from Anopheles culicifacies for epitope design using Immuno-informatics models. Bioinformation 2019; 15:600-612. [PMID: 31719771 PMCID: PMC6822521 DOI: 10.6026/97320630015600] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 09/10/2019] [Indexed: 11/23/2022] Open
Abstract
Analysis of the Amino-peptidase N (APN) protein from Anopheles culicifacies as a vector based Transmission Blocking Vaccines (TBV) target has been considered for malaria vaccine development. Short peptides as potential epitopes for B cells and cytotoxic T cells and/or helper T cells were identified using prediction models provided by NetCTL and IEDB servers. Antigenicity determination, allergenicity, immunogenicity, epitope conservancy analysis, atomic interaction with HLA allele specific structure models and population coverage were investigated in this study. The analysis of the target protein helped to identify conserved regions as potential epitopes of APN in various Anopheles species. The T cell epitopes like peptides were further analyzed by using molecular docking to check interactions against the allele specific HLA models. Thus, we report the predicted B cell (VDERYRL) and T cell (RRYLATTQF for HLA class I and LKATFTVSI for HLA class II) epitopes like peptides from APN protein of Anopheles culicifacies (Diptera: Culicidae) for further consideration as vaccine candidates subsequent to in vitro and in vivo analysis.
Collapse
Affiliation(s)
- Renu Jakhar
- Centre for Medical Biotechnology, Maharshi Dayanand University, Rohtak - 124001, Haryana
| | - Pawan Kumar
- Centre for Bioinformatics, Maharshi Dayanand University, Rohtak - 124001, Haryana
| | - Neelam Sehrawat
- Department of Genetics, Maharshi Dayanand University, Rohtak - 124001, Haryana
| | - Surendra Kumar Gakhar
- Centre for Medical Biotechnology, Maharshi Dayanand University, Rohtak - 124001, Haryana
| |
Collapse
|
31
|
Wetzel D, Chan JA, Suckow M, Barbian A, Weniger M, Jenzelewski V, Reiling L, Richards JS, Anderson DA, Kouskousis B, Palmer C, Hanssen E, Schembecker G, Merz J, Beeson JG, Piontek M. Display of malaria transmission-blocking antigens on chimeric duck hepatitis B virus-derived virus-like particles produced in Hansenula polymorpha. PLoS One 2019; 14:e0221394. [PMID: 31483818 PMCID: PMC6726142 DOI: 10.1371/journal.pone.0221394] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 08/07/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Malaria caused by Plasmodium falciparum is one of the major threats to human health globally. Despite huge efforts in malaria control and eradication, highly effective vaccines are urgently needed, including vaccines that can block malaria transmission. Chimeric virus-like particles (VLP) have emerged as a promising strategy to develop new malaria vaccine candidates. METHODS We developed yeast cell lines and processes for the expression of malaria transmission-blocking vaccine candidates Pfs25 and Pfs230 as VLP and VLP were analyzed for purity, size, protein incorporation rate and expression of malaria antigens. RESULTS In this study, a novel platform for the display of Plasmodium falciparum antigens on chimeric VLP is presented. Leading transmission-blocking vaccine candidates Pfs25 and Pfs230 were genetically fused to the small surface protein (dS) of the duck hepatitis B virus (DHBV). The resulting fusion proteins were co-expressed in recombinant Hansenula polymorpha (syn. Pichia angusta, Ogataea polymorpha) strains along with the wild-type dS as the VLP scaffold protein. Through this strategy, chimeric VLP containing Pfs25 or the Pfs230-derived fragments Pfs230c or Pfs230D1M were purified. Up to 100 mg chimeric VLP were isolated from 100 g dry cell weight with a maximum protein purity of 90% on the protein level. Expression of the Pfs230D1M construct was more efficient than Pfs230c and enabled VLP with higher purity. VLP showed reactivity with transmission-blocking antibodies and supported the surface display of the malaria antigens on the native VLP. CONCLUSION The incorporation of leading Plasmodium falciparum transmission-blocking antigens into the dS-based VLP scaffold is a promising novel strategy for their display on nano-scaled particles. Competitive processes for efficient production and purification were established in this study.
Collapse
Affiliation(s)
- David Wetzel
- ARTES Biotechnology GmbH, Langenfeld, Germany
- Laboratory of Plant and Process Design, Technical University of Dortmund, Dortmund, Germany
| | - Jo-Anne Chan
- Burnet Institute for Medical Research and Public Health, Melbourne, Victoria, Australia
| | | | - Andreas Barbian
- Düsseldorf University Hospital, Institute for Anatomy I, Düsseldorf, Germany
| | | | | | - Linda Reiling
- Burnet Institute for Medical Research and Public Health, Melbourne, Victoria, Australia
| | - Jack S. Richards
- Burnet Institute for Medical Research and Public Health, Melbourne, Victoria, Australia
| | - David A. Anderson
- Burnet Institute for Medical Research and Public Health, Melbourne, Victoria, Australia
| | - Betty Kouskousis
- Burnet Institute for Medical Research and Public Health, Melbourne, Victoria, Australia
| | - Catherine Palmer
- Burnet Institute for Medical Research and Public Health, Melbourne, Victoria, Australia
| | - Eric Hanssen
- The Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Melbourne, Victoria, Australia
| | - Gerhard Schembecker
- Laboratory of Plant and Process Design, Technical University of Dortmund, Dortmund, Germany
| | - Juliane Merz
- Evonik Technology & Infrastructure GmbH, Hanau, Germany
| | - James G. Beeson
- Burnet Institute for Medical Research and Public Health, Melbourne, Victoria, Australia
- Central Clinical School and Department of Microbiology, Monash University, Melbourne, Victoria, Australia
- Department of Medicine, Royal Melbourne Hospital, University of Melbourne, Melbourne, Victoria, Australia
| | | |
Collapse
|
32
|
Chaves LB, Perce-da-Silva DDS, Totino PRR, Riccio EKP, Baptista BDO, de Souza ABL, Rodrigues-da-Silva RN, Machado RLD, de Souza RM, Daniel-Ribeiro CT, Banic DM, Pratt-Riccio LR, Lima-Junior JDC. Plasmodium vivax ookinete surface protein (Pvs25) is highly conserved among field isolates from five different regions of the Brazilian Amazon. INFECTION GENETICS AND EVOLUTION 2019; 73:287-294. [DOI: 10.1016/j.meegid.2019.05.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 05/03/2019] [Accepted: 05/04/2019] [Indexed: 12/29/2022]
|
33
|
Zhu X, Sun L, He Y, Wei H, Hong M, Liu F, Liu Q, Cao Y, Cui L. Plasmodium berghei serine/threonine protein phosphatase PP5 plays a critical role in male gamete fertility. Int J Parasitol 2019; 49:685-695. [DOI: 10.1016/j.ijpara.2019.03.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Accepted: 03/18/2019] [Indexed: 02/06/2023]
|
34
|
Miura K, Swihart BJ, Deng B, Zhou L, Pham TP, Diouf A, Fay MP, Long CA. Strong concordance between percent inhibition in oocyst and sporozoite intensities in a Plasmodium falciparum standard membrane-feeding assay. Parasit Vectors 2019; 12:206. [PMID: 31060594 PMCID: PMC6501457 DOI: 10.1186/s13071-019-3470-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 04/29/2019] [Indexed: 11/22/2022] Open
Abstract
Background Effective malaria transmission-blocking vaccines (TBVs) can support malaria eradication programmes, and the standard membrane-feeding assay (SMFA) has been used as a “gold standard” assay for TBV development. However, in SMFA, the inhibitory activity is commonly measured at oocyst stage of parasites, while it is the sporozoites which transmit malaria from a mosquito to a human. A handful of studies have shown that there is a positive correlation between oocyst and sporozoite intensities. However, no study has been completed to compare inhibition levels in oocyst and sporozoite intensities in the presence of transmission-blocking (TB) antibodies. Results Plasmodium falciparum NF54 gametocytes were fed to Anopheles stephensi mosquitoes with or without anti-Pfs25 or anti-Pfs48/45 TB antibodies in 15 independent assays. For each group, a portion of the mosquitoes was dissected for oocyst counts (day 8 after feed), and a portion of the remaining mosquitoes was dissected for sporozoite counts (day 16). This study covered a large range of oocyst and sporozoite intensities: 0.2 to 80.5 on average for oocysts, and 141 to 77,417 for sporozoites. The sporozoite data were well explained by a zero-inflated negative binomial model, regardless of the presence or absence of TB antibodies. Inhibition levels in both oocyst and sporozoite intensities were determined within the same groups in 9 independent assays. When the level of inhibition in sporozoite number (expressed as Log Mean Ratio, LMR; average number in a control group was divided by the one in a test group, then took a log of the ratio) was plotted against LMR in oocyst number, the best-fit slope of a linear regression was not different from 1 (the best estimate, 1.08; 95% confidence interval, 0.87 to 1.29). Furthermore, a Bland–Altman analysis showed a strong agreement between inhibitions in oocysts and in sporozoites. Conclusions The results indicate that percent inhibition in oocyst intensity of a test sample can be directly converted to % inhibition in sporozoite intensity in P. falciparum SMFA. Therefore, if sporozoite intensity determines transmission rate from mosquitoes to humans, the percent inhibition in oocyst intensity measured by SMFA can be used to estimate the TBV efficacy. Electronic supplementary material The online version of this article (10.1186/s13071-019-3470-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- 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.
| | - Bruce J Swihart
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 5601 Fishers Lane, Rockville, MD, 20852, USA
| | - Bingbing Deng
- 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
| | - Thao P Pham
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway, Rockville, MD, 20852, USA
| | - Ababacar Diouf
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway, Rockville, MD, 20852, USA
| | - Michael P Fay
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 5601 Fishers Lane, Rockville, MD, 20852, 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
| |
Collapse
|
35
|
Miura K, Deng B, Wu Y, Zhou L, Pham TP, Diouf A, Wu CK, Lee SM, Plieskatt JL, Morin MJ, Long CA. ELISA units, IgG subclass ratio and avidity determined functional activity of mouse anti-Pfs230 antibodies judged by a standard membrane-feeding assay with Plasmodium falciparum. Vaccine 2019; 37:2073-2078. [PMID: 30850239 DOI: 10.1016/j.vaccine.2019.02.071] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 02/22/2019] [Accepted: 02/24/2019] [Indexed: 10/27/2022]
Abstract
The standard membrane-feeding assay (SMFA) is a functional assay that has been used to inform the development of transmission-blocking vaccines (TBV) against Plasmodium falciparum malaria. For Pfs230, a lead target antigen for TBV development, a few studies have tested either a single anti-Pfs230 polyclonal or monoclonal antibody (one antibody per study) at serial dilutions and showed a dose-dependent response. Further, there have been reports that the SMFA activity of anti-Pfs230 polyclonal and monoclonal antibodies were enhanced in the presence of complement. However, no analysis has been performed with multiple samples, and the impact of anti-Pfs230 antibody titers, IgG subclass profile and avidity were evaluated together in relation to transmission-reducing activity (TRA) by SMFA. In this report, a total of 39 unique anti-Pfs230 IgGs from five different mouse immunization studies were assessed for their ELISA units (EU), IgG2/IgG1 ratio and avidity by ELISA, and the functionality (% transmission-reducing activity, %TRA) by SMFA. The mice were immunized with Pfs230 alone, Pfs230 conjugated to CRM197, or a mixture of unconjugated Pfs230 and CRM197 proteins using Alhydrogel or Montanide ISA720 adjuvants. In all studies, the Pfs230 antigen was from the same source. There was a significant correlation between EU and %TRA (p < 0.0001 by a Spearman rank test) for the anti-Pfs230 IgGs. Notably, multiple linear regression analyses showed that both IgG2/IgG1 ratio and avidity significantly affected %TRA (p = 0.003 to p = 0.014, depending on the models) after adjusting for EU. The results suggest that in addition to antibody titers, IgG2/IgG1 ratio and avidity should each be evaluated to predict the biological activity of anti-Pfs230 antibodies for future vaccine development.
Collapse
Affiliation(s)
- Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA.
| | - Bingbing Deng
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Yimin Wu
- PATH's Malaria Vaccine Initiative (MVI), Washington, DC 20001, USA
| | - Luwen Zhou
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Thao P Pham
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Ababacar Diouf
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Chia-Kuei Wu
- PATH's Malaria Vaccine Initiative (MVI), Washington, DC 20001, USA
| | - Shwu-Maan Lee
- PATH's Malaria Vaccine Initiative (MVI), Washington, DC 20001, 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
| |
Collapse
|
36
|
Identification of domains within Pfs230 that elicit transmission blocking antibody responses. Vaccine 2019; 37:1799-1806. [PMID: 30824357 PMCID: PMC6708081 DOI: 10.1016/j.vaccine.2019.02.021] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 01/23/2019] [Accepted: 02/05/2019] [Indexed: 11/23/2022]
Abstract
A transmission-blocking vaccine (TBV) against Plasmodium falciparum is likely to be a valuable tool in a malaria eradication program. Pfs230 is one of the major TBV candidates, and multiple Pfs230-based vaccines induced antibodies, which prevented oocyst formation in mosquitoes as determined by a standard membrane-feeding assay (SMFA). Pfs230 is a >300 kDa protein consisting of 14 cysteine motif (CM) domains, and the size and cysteine-rich nature of the molecule have hampered its production as an intact protein. Except for one early study with maltose-binding protein fusion Pfs230 constructs expressed in Esherichia coli, all other studies have focused on only the first four CM domains in the Pfs230 molecule. To identify all possible TBV candidate domains, we systematically produced either single-CM-domain (a total of 14), 2-CM-domain (7), or 4-CM-domain (6) recombinant protein fragments using a eukaryotic wheat germ cell-free expression system (WGCFS). In addition, two more constructs which covered previously published regions, and an N-terminal prodomain construct spanning the natural cleavage site of Pfs230 were produced. Antisera against each fragment were generated in mice and we evaluated the reactivity to native Pfs230 protein by Western blots and immunofluorescence assay (IFA), and functionality by SMFA. All 30 WGCFS-produced Pfs230 constructs were immunogenic in mice. Approximately half of the mouse antibodies specifically recognized native Pfs230 by Western blots with variable band intensities. Among them, seven antibodies showed higher reactivities against native Pfs230 determined by IFA. Interestingly, antibodies against all protein fragments containing CM domain 1 displayed strong inhibitions in SMFA, while antibodies generated using constructs without CM domain 1 showed no inhibition. The results strongly support the concept that future Pfs230-based vaccine development should focus on the Pfs230 CM domain 1.
Collapse
|
37
|
Habtewold T, Tapanelli S, Masters EKG, Hoermann A, Windbichler N, Christophides GK. Streamlined SMFA and mosquito dark-feeding regime significantly improve malaria transmission-blocking assay robustness and sensitivity. Malar J 2019; 18:24. [PMID: 30683107 PMCID: PMC6347765 DOI: 10.1186/s12936-019-2663-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 01/19/2019] [Indexed: 02/03/2023] Open
Abstract
Background The development of malaria transmission-blocking strategies including the generation of malaria refractory mosquitoes to replace the wild populations through means of gene drives hold great promise. The standard membrane feeding assay (SMFA) that involves mosquito feeding on parasitized blood through an artificial membrane system is a vital tool for evaluating the efficacy of transmission-blocking interventions. However, despite the availability of several published protocols, the SMFA remains highly variable and broadly insensitive. Methods The SMFA protocol was optimized through coordinated culturing of Anopheles coluzzii mosquitoes and Plasmodium falciparum parasite coupled with placing mosquitoes under a strict dark regime before, during, and after the gametocyte feed. Results A detailed description of essential steps is provided toward synchronized generation of highly fit An. coluzzii mosquitoes and P. falciparum gametocytes in preparation for an SMFA. A dark-infection regime that emulates the natural vector-parasite interaction system is described, which results in a significant increase in the infection intensity and prevalence. Using this optimal SMFA pipeline, a series of putative transmission-blocking antimicrobial peptides (AMPs) were screened, confirming that melittin and magainin can interfere with P. falciparum development in the vector. Conclusion A robust SMFA protocol that enhances the evaluation of interventions targeting human malaria transmission in laboratory setting is reported. Melittin and magainin are identified as highly potent antiparasitic AMPs that can be used for the generation of refractory Anopheles gambiae mosquitoes.
Collapse
Affiliation(s)
- Tibebu Habtewold
- Department of Life Sciences, Imperial College London, London, UK.
| | - Sofia Tapanelli
- Department of Life Sciences, Imperial College London, London, UK
| | | | - Astrid Hoermann
- Department of Life Sciences, Imperial College London, London, UK
| | | | | |
Collapse
|
38
|
Coelho CH, Gazzinelli-Guimaraes PH, Howard J, Barnafo E, Alani NAH, Muratova O, McCormack A, Kelnhofer E, Urban JF, Narum DL, Anderson C, Langhorne J, Nutman TB, Duffy PE. Chronic helminth infection does not impair immune response to malaria transmission blocking vaccine Pfs230D1-EPA/Alhydrogel® in mice. Vaccine 2019; 37:1038-1045. [PMID: 30685251 PMCID: PMC6382667 DOI: 10.1016/j.vaccine.2019.01.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 01/08/2019] [Accepted: 01/09/2019] [Indexed: 12/20/2022]
Abstract
Pfs230 is a candidate malaria transmission blocking vaccine against P. falciparum. Pfs230 vaccine is being tested in areas where malaria and helminth infections are co-endemic. Chronic helminth infection induces a marked increase in systemic Th2 and regulatory cytokine levels in mice. Chronic H. polygyrus bakeri infection does not alter Pfs230 vaccine specific-antibody levels. Functional activity of Pfs230 vaccine was not impaired by chronic helminth infection in mice.
Introduction Malaria transmission blocking vaccines (TBV) are innovative approaches that aim to induce immunity in humans against Plasmodium during mosquito stage, neutralizing the capacity of the infected vectors to transmit malaria. Pfs230D1-EPA/Alhydrogel®, a promising protein-protein conjugate malaria TBV, is currently being tested in human clinical trials in areas where P. falciparum malaria is coendemic with helminth parasites. Helminths are complex metazoans that share the master capacity to downregulate the host immune response towards themselves and also to bystander antigens, including vaccines. However, it is not known whether the activity of a protein-based malaria TBV may be affected by a chronic helminth infection. Methods Using an experimental murine model for a chronic helminth infection (Heligmosomoides polygyrus bakeri - Hpb), we evaluated whether prior infection alters the activity of Pfs230D1-EPA/Alhydrogel® TBV in mice. Results After establishment of a chronic infection, characterized by a marked increase of parasite antigen-specific IgG1, IgA and IgE antibody responses, concomitant with an increase of systemic IL-10, IL-5 and IL-6 levels, the Hpb-infected mice were immunized with Pfs230D1-EPA/Alhydrogel® and the vaccine-specific immune response was compared with that in non-infected immunized mice. TBV immunizations induced an elevated vaccine specific-antibody response, however Pfs230D1 specific-IgG levels were similar between infected and uninfected mice at days 15, 25 and 35 post-vaccination. Absolute numbers of Pfs230D1-activated B cells generated in response to the vaccine were also similar among the vaccinated groups. Finally, vaccine activity assessed by reduction of oocyst number in P. falciparum infected mosquitoes was similar between Hpb-infected and immunized mice with non-infected immunized mice. Conclusion Pfs230D1-EPA/Alhydrogel® efficacy is not impaired by a chronic helminth infection in mice.
Collapse
Affiliation(s)
- Camila H Coelho
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | | | - Jennifer Howard
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Emma Barnafo
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Nada A H Alani
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Olga Muratova
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Ashley McCormack
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Emily Kelnhofer
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Joseph F Urban
- US Department of Agriculture, Agricultural Research Service, Beltsville Human Nutrition Research Center, Diet, Genomic and Immunology Laboratory, Beltsville, MD, USA
| | - David L Narum
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Charles Anderson
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | | | - Thomas B Nutman
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Patrick E Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA.
| |
Collapse
|
39
|
Beeson JG, Kurtovic L, Dobaño C, Opi DH, Chan JA, Feng G, Good MF, Reiling L, Boyle MJ. Challenges and strategies for developing efficacious and long-lasting malaria vaccines. Sci Transl Med 2019; 11:11/474/eaau1458. [DOI: 10.1126/scitranslmed.aau1458] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 08/05/2018] [Accepted: 11/02/2018] [Indexed: 12/24/2022]
Abstract
Although there has been major recent progress in malaria vaccine development, substantial challenges remain for achieving highly efficacious and durable vaccines against Plasmodium falciparum and Plasmodium vivax malaria. Greater knowledge of mechanisms and key targets of immunity are needed to accomplish this goal, together with new strategies for generating potent, long-lasting, functional immunity against multiple antigens. Implementation considerations in endemic areas will ultimately affect vaccine effectiveness, so innovations to simplify and enhance delivery are also needed. Whereas challenges remain, recent exciting progress and emerging knowledge promise hope for the future of malaria vaccines.
Collapse
|
40
|
Tachibana M, Ishino T, Tsuboi T, Torii M. The Plasmodium yoelii microgamete surface antigen (PyMiGS) induces anti-malarial transmission blocking immunity that reduces microgamete motility/release from activated male gametocytes. Vaccine 2018; 36:7463-7471. [DOI: 10.1016/j.vaccine.2018.10.067] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Revised: 10/17/2018] [Accepted: 10/19/2018] [Indexed: 02/03/2023]
|
41
|
Cockburn IA, Seder RA. Malaria prevention: from immunological concepts to effective vaccines and protective antibodies. Nat Immunol 2018; 19:1199-1211. [PMID: 30333613 DOI: 10.1038/s41590-018-0228-6] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 08/31/2018] [Indexed: 02/08/2023]
Abstract
Development of a malaria vaccine remains a critical priority to decrease clinical disease and mortality and facilitate eradication. Accordingly, RTS,S, a protein-subunit vaccine, has completed phase III clinical trials and confers ~30% protection against clinical infection over 4 years. Whole-attenuated-sporozoite and viral-subunit vaccines induce between 20% and 100% protection against controlled human malaria infection, but there is limited published evidence to date for durable, high-level efficacy (>50%) against natural exposure. Importantly, fundamental scientific advances related to the potency, durability, breadth and location of immune responses will be required for improving vaccine efficacy with these and other vaccine approaches. In this Review, we focus on the current understanding of immunological mechanisms of protection from animal models and human vaccine studies, and on how these data should inform the development of next-generation vaccines. Furthermore, we introduce the concept of using passive immunization with monoclonal antibodies as a new approach to prevent and eliminate malaria.
Collapse
Affiliation(s)
- Ian A Cockburn
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Robert A Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
42
|
The Plasmodium falciparum male gametocyte protein P230p, a paralog of P230, is vital for ookinete formation and mosquito transmission. Sci Rep 2018; 8:14902. [PMID: 30297725 PMCID: PMC6175877 DOI: 10.1038/s41598-018-33236-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 09/25/2018] [Indexed: 12/22/2022] Open
Abstract
Two members of 6-cysteine (6-cys) protein family, P48/45 and P230, are important for gamete fertility in rodent and human malaria parasites and are leading transmission blocking vaccine antigens. Rodent and human parasites encode a paralog of P230, called P230p. While P230 is expressed in male and female parasites, P230p is expressed only in male gametocytes and gametes. In rodent malaria parasites this protein is dispensable throughout the complete life-cycle; however, its function in P. falciparum is unknown. Using CRISPR/Cas9 methodology we disrupted the gene encoding Pfp230p resulting in P. falciparum mutants (PfΔp230p) lacking P230p expression. The PfΔp230p mutants produced normal numbers of male and female gametocytes, which retained expression of P48/45 and P230. Upon activation male PfΔp230p gametocytes undergo exflagellation and form male gametes. However, male gametes are unable to attach to red blood cells resulting in the absence of characteristic exflagellation centres in vitro. In the absence of P230p, zygote formation as well as oocyst and sporozoite development were strongly reduced (>98%) in mosquitoes. These observations demonstrate that P230p, like P230 and P48/45, has a vital role in P. falciparum male fertility and zygote formation and warrants further investigation as a potential transmission blocking vaccine candidate.
Collapse
|
43
|
Reed SG, Carter D, Casper C, Duthie MS, Fox CB. Correlates of GLA family adjuvants' activities. Semin Immunol 2018; 39:22-29. [PMID: 30366662 PMCID: PMC6289613 DOI: 10.1016/j.smim.2018.10.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 10/03/2018] [Indexed: 12/30/2022]
Abstract
Lipopolysaccharide (LPS) is a well-defined agonist of Toll-like receptor (TLR) 4 that activates innate immune responses and influences the development of the adaptive response during infection with Gram-negative bacteria. Many years ago, Dr. Edgar Ribi separated the adjuvant activity of LPS from its toxic effects, an effort that led to the development of monophosphoryl lipid A (MPL). MPL, derived from Salmonella minnesota R595, has progressed through clinical development and is now used in various product-enabling formulations to support the generation of antigen-specific responses in several commercial and preclinical vaccines. We have generated several synthetic lipid A molecules, foremost glucopyranosyl lipid adjuvant (GLA) and second-generation lipid adjuvant (SLA), and have advanced these to clinical trial for various indications. In this review we summarize the potential and current positioning of TLR4-based adjuvant formulations in approved and emerging vaccines.
Collapse
Affiliation(s)
- Steven G Reed
- Infectious Disease Research Institute, 1616 Eastlake Ave E, Suite 400, Seattle, WA 98102 USA.
| | - Darrick Carter
- Infectious Disease Research Institute, 1616 Eastlake Ave E, Suite 400, Seattle, WA 98102 USA.
| | - Corey Casper
- Infectious Disease Research Institute, 1616 Eastlake Ave E, Suite 400, Seattle, WA 98102 USA.
| | - Malcolm S Duthie
- Infectious Disease Research Institute, 1616 Eastlake Ave E, Suite 400, Seattle, WA 98102 USA.
| | - Christopher B Fox
- Infectious Disease Research Institute, 1616 Eastlake Ave E, Suite 400, Seattle, WA 98102 USA.
| |
Collapse
|
44
|
Antimalarial Transmission-Blocking Interventions: Past, Present, and Future. Trends Parasitol 2018; 34:735-746. [PMID: 30082147 DOI: 10.1016/j.pt.2018.07.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 06/14/2018] [Accepted: 07/02/2018] [Indexed: 12/17/2022]
Abstract
Malaria remains a major global health challenge. Appropriate use of current antimalarial tools has reduced the disease burden, but morbidity and mortality remain unacceptably high. It is widely accepted that, to achieve long-term control/eradication, it will be necessary to use interventions that inhibit the transmission of parasites to mosquitoes - these tools are termed transmission-blocking interventions (TBIs). This article aims to outline the rationale for the development of TBIs, with a focus on transmission-blocking drugs and (parasite-derived) transmission-blocking vaccines. We describe and summarise the current status of each of these intervention classes and attempt to identify future requirements in development, with a focus on the challenges of establishing each method within an integrated malarial control programme in the future.
Collapse
|
45
|
Ishino T, Tsuboi T. Progress toward a transmission-blocking vaccine against malaria. THE LANCET. INFECTIOUS DISEASES 2018; 18:927-928. [PMID: 30061052 DOI: 10.1016/s1473-3099(18)30358-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 05/29/2018] [Indexed: 11/17/2022]
Affiliation(s)
- Tomoko Ishino
- Division of Molecular Parasitology, 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
|
46
|
Characterization of Plasmodium berghei Pbg37 as Both a Pre- and Postfertilization Antigen with Transmission-Blocking Potential. Infect Immun 2018; 86:IAI.00785-17. [PMID: 29866905 DOI: 10.1128/iai.00785-17] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 05/26/2018] [Indexed: 01/29/2023] Open
Abstract
Transmission-blocking vaccines (TBVs) interrupting malaria transmission are an integrated tool for malaria eradication. We characterized a sexual-stage-specific gene (PBANKA_060330) from Plasmodium berghei and studied its potential for use as a TBV. This gene, referred to as pbg37, encodes a protein of 37 kDa with a signal peptide and multiple transmembrane domains and is preferentially expressed in gametocytes. A recombinant Pbg37 (rPbg37) protein targeting the N-terminal 63 amino acids (amino acids 26 to 88) expressed in bacteria elicited strong antibody responses in mice. Western blotting demonstrated Pbg37 expression in gametocytes, zygotes, and, to a lesser extent, ookinetes and its predominant association with the membranes of gametocytes. Indirect immunofluorescence assay showed an abundant surface localization of Pbg37 on gametes and zygotes but reduced amounts on retorts and ookinetes. Knockout of pbg37 (Δpbg37) led to a considerable reduction in gametocytemia, which translated into a ~92.1% decrease in the oocyst number in mosquitoes. Deletion of pbg37 had a more substantial influence on the development and maturation of microgametocytes. As a result, the Δpbg37 lines exhibited a higher female/male gametocyte ratio, fewer mature male gametocytes, and defects in the exflagellation of mature microgametocytes. To test the transmission-blocking potential of Pbg37, an in vitro ookinete assay showed that the major inhibitory effects of anti-Pbg37 antiserum were on the exflagellation and fertilization processes. Direct feeding of mosquitoes on mice immunized with rPbg37 or a control protein showed that rPbg37-immunized and P. berghei-infected mice had a significant reduction (49.1%) in oocyst density compared to the controls. The conservation of this gene in Plasmodium warrants further investigations in human malaria parasites.
Collapse
|
47
|
Swihart BJ, Fay MP, Miura K. Statistical Methods for Standard Membrane-Feeding Assays to Measure Transmission Blocking or Reducing Activity in Malaria. J Am Stat Assoc 2018; 113:534-545. [PMID: 31007315 DOI: 10.1080/01621459.2017.1356313] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Transmission blocking vaccines for malaria are not designed to directly protect vaccinated people from malaria disease, but to reduce the probability of infecting other people by interfering with the growth of the malaria parasite in mosquitoes. Standard membrane-feeding assays compare the growth of parasites in mosquitoes from a test sample (using antibodies from a vaccinated person) compared to a control sample. There is debate about whether to estimate the transmission reducing activity (TRA) which compares the mean number of parasites between test and control samples, or transmission blocking activity (TBA) which compares the proportion of infected mosquitoes. TBA appears biologically more important since each mosquito with any parasites is potentially infective; however, TBA is less reproducible and may be an overly strict criterion for screening vaccine candidates. Through a statistical model, we show that the TBA estimand depends on μ c , the mean number of parasites in the control mosquitoes, a parameter not easily experimentally controlled. We develop a standardized TBA estimator based on the model and a given target value for μ c which has better mean squared error than alternative methods. We discuss types of statistical inference needed for using these assays for vaccine development.
Collapse
Affiliation(s)
| | - Michael P Fay
- Biostatistics Research Branch, NIAID, NIH, Rockville, MD
| | - Kazutoyo Miura
- National Institute of Allergy and Infectious Diseases, Laboratory of Malaria and Vector Research, National Institutes of Health, Rockville, MD
| |
Collapse
|
48
|
Tachibana M, Ishino T, Takashima E, Tsuboi T, Torii M. A male gametocyte osmiophilic body and microgamete surface protein of the rodent malaria parasite Plasmodium yoelii (PyMiGS) plays a critical role in male osmiophilic body formation and exflagellation. Cell Microbiol 2018; 20:e12821. [PMID: 29316140 PMCID: PMC5901010 DOI: 10.1111/cmi.12821] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 12/15/2017] [Accepted: 12/22/2017] [Indexed: 01/18/2023]
Abstract
Anopheles mosquitoes transmit Plasmodium parasites of mammals, including the species that cause malaria in humans. Malaria pathology is caused by rapid multiplication of parasites in asexual intraerythrocytic cycles. Sexual stage parasites are also produced during the intraerythrocytic cycle and are ingested by the mosquito, initiating gametogenesis and subsequent sporogonic stage development. Here, we present a Plasmodium protein, termed microgamete surface protein (MiGS), which has an important role in male gametocyte osmiophilic body (MOB) formation and microgamete function. MiGS is expressed exclusively in male gametocytes and microgametes, in which MiGS localises to the MOB and microgamete surface. Targeted gene disruption of MiGS in a rodent malaria parasite Plasmodium yoelii 17XNL generated knockout parasites (ΔPyMiGS) that proliferate normally in erythrocytes and form male and female gametocytes. The number of MOB in male gametocyte cytoplasm is markedly reduced and the exflagellation of microgametes is impaired in ΔPyMiGS. In addition, anti‐PyMiGS antibody severely blocked the parasite development in the Anopheles stephensi mosquito. MiGS might thus be a potential novel transmission‐blocking vaccine target candidate.
Collapse
Affiliation(s)
- Mayumi Tachibana
- Division of Molecular Parasitology, Proteo-Science Center, Ehime University, Toon, Ehime, Japan
| | - Tomoko Ishino
- Division of Molecular Parasitology, Proteo-Science Center, Ehime University, Toon, Ehime, Japan
| | - Eizo Takashima
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Ehime, Japan
| | - Takafumi Tsuboi
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Ehime, Japan
| | - Motomi Torii
- Division of Molecular Parasitology, Proteo-Science Center, Ehime University, Toon, Ehime, Japan
| |
Collapse
|
49
|
|
50
|
Yamamoto DS, Sumitani M, Hatakeyama M, Matsuoka H. Malaria infectivity of xanthurenic acid-deficient anopheline mosquitoes produced by TALEN-mediated targeted mutagenesis. Transgenic Res 2018; 27:51-60. [DOI: 10.1007/s11248-018-0057-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 01/11/2018] [Indexed: 12/15/2022]
|