1
|
Bhalerao P, Singh S, Prajapati VK, Bhatt TK. Exploring malaria parasite surface proteins to devise highly immunogenic multi-epitope subunit vaccine for Plasmodium falciparum. J Genet Eng Biotechnol 2024; 22:100377. [PMID: 38797552 PMCID: PMC11089370 DOI: 10.1016/j.jgeb.2024.100377] [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: 12/12/2023] [Revised: 03/07/2024] [Accepted: 04/13/2024] [Indexed: 05/29/2024]
Abstract
BACKGROUND Malaria has remained a major health concern for decades among people living in tropical and sub-tropical countries. Plasmodium falciparum is one of the critical species that cause severe malaria and is responsible for major mortality. Moreover, the parasite has generated resistance against all WHO recommended drugs and therapies. Therefore, there is an urgent need for preventive measures in the form of reliable vaccines to achieve the target of a malaria-free world. Surface proteins are the preferable choice for subunit vaccine development because they are rapidly detected and engaged by host immune cells and vaccination-induced antibodies. Additionally, abundant surface or membrane proteins may contribute to the opsonization of pathogens by vaccine-induced antibodies. RESULTS In our study, we have listed all those surface proteins from the literature that could be functionally important and essential for infection and immune evasion of the malaria parasite. Eight Plasmodium surface and membrane proteins from the pre-erythrocyte and erythrocyte stages were shortlisted. Thirty-seven epitopes (B-cell, CTL, and HTL epitopes) from these proteins were predicted using immune-informatic tools and joined with suitable peptide linkers to design a vaccine construct. A TLR-4 agonist peptide adjuvant was added at the N-terminus of the multi-epitope series, followed by the PADRE sequence and EAAAK linker. The TLR-4 receptor was docked with the construct's anticipated model structure. The complex of vaccine and TLR-4, with the lowest energy -1514, was found to be stable under simulated physiological settings. CONCLUSION This study has provided a novel multi-epitope construct that may be exploited further for the development of an efficient vaccine for malaria.
Collapse
Affiliation(s)
- Preshita Bhalerao
- Department of Biotechnology, School of Life Sciences, Central University of Rajasthan, Bandar Sindri, Kishangarh, Ajmer 305817, Rajasthan, India
| | - Satyendra Singh
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Bandar Sindri, Kishangarh, Ajmer 305817, Rajasthan, India
| | - Vijay Kumar Prajapati
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi 110021, India
| | - Tarun Kumar Bhatt
- Department of Biotechnology, School of Life Sciences, Central University of Rajasthan, Bandar Sindri, Kishangarh, Ajmer 305817, Rajasthan, India.
| |
Collapse
|
2
|
Tandoh KZ, Ibarra-Meneses AV, Langlais D, Olivier M, Torrecilhas AC, Fernandez-Prada C, Regev-Rudzki N, Duah-Quashie NO. Extracellular Vesicles: Translational Agenda Questions for Three Protozoan Parasites. Traffic 2024; 25:e12935. [PMID: 38629580 DOI: 10.1111/tra.12935] [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: 10/16/2023] [Revised: 04/02/2024] [Accepted: 04/03/2024] [Indexed: 04/19/2024]
Abstract
The protozoan parasites Plasmodium falciparum, Leishmania spp. and Trypanosoma cruzi continue to exert a significant toll on the disease landscape of the human population in sub-Saharan Africa and Latin America. Control measures have helped reduce the burden of their respective diseases-malaria, leishmaniasis and Chagas disease-in endemic regions. However, the need for new drugs, innovative vaccination strategies and molecular markers of disease severity and outcomes has emerged because of developing antimicrobial drug resistance, comparatively inadequate or absent vaccines, and a lack of trustworthy markers of morbid outcomes. Extracellular vesicles (EVs) have been widely reported to play a role in the biology and pathogenicity of P. falciparum, Leishmania spp. and T. cruzi ever since they were discovered. EVs are secreted by a yet to be fully understood mechanism in protozoans into the extracellular milieu and carry a cargo of diverse molecules that reflect the originator cell's metabolic state. Although our understanding of the biogenesis and function of EVs continues to deepen, the question of how EVs in P. falciparum, Leishmania spp. and T. cruzi can serve as targets for a translational agenda into clinical and public health interventions is yet to be fully explored. Here, as a consortium of protozoan researchers, we outline a plan for future researchers and pose three questions to direct an EV's translational agenda in P. falciparum, Leishmania spp. and T. cruzi. We opine that in the long term, executing this blueprint will help bridge the current unmet needs of these medically important protozoan diseases in sub-Saharan Africa and Latin America.
Collapse
Affiliation(s)
- Kwesi Z Tandoh
- West African Centre for Cell Biology of Infectious Pathogens, Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
- Department of Epidemiology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Ana Victoria Ibarra-Meneses
- Département de Pathologie et Microbiologie, Faculté de Médecine Vétérinaire, Université de Montréal, Montreal, Canada
- The Research Group on Infectious Diseases in Production Animals (GREMIP), Faculty of Veterinary Medicine, Université de Montréal, Montreal, Canada
| | - David Langlais
- Department of Human Genetics, Dahdaleh Institute of Genomic Medicine, Montreal, Canada
- Department of Microbiology and Immunology, McGill Research Centre on Complex Traits, Montreal, Canada
| | - Martin Olivier
- Department of Microbiology and Immunology, McGill Research Centre on Complex Traits, Montreal, Canada
- IDIGH, The Research Institute of the McGill University Health Centre, McGill University, Montreal, Canada
| | - Ana Claudia Torrecilhas
- Departamento de Ciências Farmacêuticas, Laboratório de Imunologia Celular e Bioquímica de Fungos e Protozoários, Universidade Federal de São Paulo (UNIFESP), Instituto de Ciências Ambientais, Químicas e Farmacêuticas, São Paulo, Brazil
| | - Christopher Fernandez-Prada
- Département de Pathologie et Microbiologie, Faculté de Médecine Vétérinaire, Université de Montréal, Montreal, Canada
- The Research Group on Infectious Diseases in Production Animals (GREMIP), Faculty of Veterinary Medicine, Université de Montréal, Montreal, Canada
- Department of Microbiology and Immunology, McGill Research Centre on Complex Traits, Montreal, Canada
| | - Neta Regev-Rudzki
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Nancy O Duah-Quashie
- Department of Epidemiology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
| |
Collapse
|
3
|
Ohene-Adjei K, Asante KP, Akuffo KO, Tounaikok N, Asiamah M, Owiredu D, Manu AA, Danso-Appiah A. Malaria vaccine-related adverse events among children under 5 in sub-Saharan Africa: systematic review and meta-analysis protocol. BMJ Open 2023; 13:e076985. [PMID: 37793915 PMCID: PMC10551995 DOI: 10.1136/bmjopen-2023-076985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 09/11/2023] [Indexed: 10/06/2023] Open
Abstract
INTRODUCTION The RTS,S vaccine has been approved for use in children under 5 living in moderate to high malaria transmission areas. However, clinically important adverse events have been reported in countries in sub-Saharan Africa. This systematic review aims to assess the frequency, severity and clinical importance of vaccine-related adverse events. METHODS AND ANALYSIS This systematic review protocol has been prepared following robust methods and reported in line with the Preferred Reporting Items for Systematic reviews and Meta-Analyses for protocols guidelines. We will search PubMed, CINAHL, LILACS, Google Scholar, SCOPUS, WEB OF SCIENCE, Cochrane library, HINARI, African Journals Online, Trip Pro and TOXNET from 2000 to 30 September 2023, without language restrictions. We will also search conference proceedings, dissertations, World Bank Open Knowledge Repository, and WHO, PATH, UNICEF, Food and Drugs Authorities and European Medicines Agency databases, preprint repositories and reference lists of relevant studies for additional studies. Experts in the field will be contacted for unpublished or published studies missed by our searches. At least two reviewers will independently select studies and extract data using pretested tools and assess risk of bias in the included studies using the Cochrane risk of bias tool. Any disagreements will be resolved through discussion between the reviewers. Heterogeneity will be explored graphically, and statistically using the I2 statistic. We will conduct random-effects meta-analysis when heterogeneity is appreciable, and express dichotomous outcomes (serious adverse events, cerebral malaria and febrile convulsion) as risk ratio (RR) with their 95% CI. We will perform subgroup analysis to assess the impact of heterogeneity and sensitivity analyses to test the robustness of the effect estimates. The overall level of evidence will be assessed using Grading of Recommendations Assessment, Development and Evaluation. ETHICS AND DISSEMINATION Ethical approval is not required for a systematic review. The findings of this study will be disseminated through stakeholder forums, conferences and peer-review publications. PROSPERO REGISTRATION NUMBER CRD42021275155.
Collapse
Affiliation(s)
- Kennedy Ohene-Adjei
- Department of Epidemiology and Disease Control, School of Public Health, College of Health Sciences, University of Ghana, Accra, Ghana
- Tain District Health Directorate, Ghana Health Service, Tain, Ghana
| | - Kwaku Poku Asante
- Research and Development Division, Kintampo Health Research Centre, Ghana Health Service, Kintampo, Kintampo North Municipality, Bono East Region, Ghana
| | - Kwadwo Owusu Akuffo
- Department of Optometry and Visual Science, College of Science, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Narcisse Tounaikok
- Centre for Evidence Synthesis and Policy, School of Public Health, University of Ghana, Accra, Ghana
- Department of Human and Animal Health, University of Emi Koussi, N'Djamena, Chad
| | - Morrison Asiamah
- Department of Electron Microscopy and Histopathology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - David Owiredu
- Department of Epidemiology and Disease Control, School of Public Health, College of Health Sciences, University of Ghana, Accra, Ghana
- Centre for Evidence Synthesis and Policy, School of Public Health, University of Ghana, Accra, Ghana
| | - Alexander Ansah Manu
- Department of Epidemiology and Disease Control, School of Public Health, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Anthony Danso-Appiah
- Department of Epidemiology and Disease Control, School of Public Health, College of Health Sciences, University of Ghana, Accra, Ghana
- Centre for Evidence Synthesis and Policy, School of Public Health, University of Ghana, Accra, Ghana
| |
Collapse
|
4
|
Su X, Stadler RV, Xu F, Wu J. Malaria Genomics, Vaccine Development, and Microbiome. Pathogens 2023; 12:1061. [PMID: 37624021 PMCID: PMC10459703 DOI: 10.3390/pathogens12081061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/14/2023] [Accepted: 08/15/2023] [Indexed: 08/26/2023] Open
Abstract
Recent advances in malaria genetics and genomics have transformed many aspects of malaria research in areas of molecular evolution, epidemiology, transmission, host-parasite interaction, drug resistance, pathogenicity, and vaccine development. Here, in addition to introducing some background information on malaria parasite biology, parasite genetics/genomics, and genotyping methods, we discuss some applications of genetic and genomic approaches in vaccine development and in studying interactions with microbiota. Genetic and genomic data can be used to search for novel vaccine targets, design an effective vaccine strategy, identify protective antigens in a whole-organism vaccine, and evaluate the efficacy of a vaccine. Microbiota has been shown to influence disease outcomes and vaccine efficacy; studying the effects of microbiota in pathogenicity and immunity may provide information for disease control. Malaria genetics and genomics will continue to contribute greatly to many fields of malaria research.
Collapse
Affiliation(s)
- Xinzhuan Su
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA; (R.V.S.); (F.X.); (J.W.)
| | | | | | | |
Collapse
|
5
|
Azmi WA, Rizki AFM, Djuardi Y, Artika IM, Siregar JE. Molecular insights into artemisinin resistance in Plasmodium falciparum: An updated review. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2023:105460. [PMID: 37269964 DOI: 10.1016/j.meegid.2023.105460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/25/2023] [Accepted: 05/27/2023] [Indexed: 06/05/2023]
Abstract
Malaria still poses a major burden on human health around the world, especially in endemic areas. Plasmodium resistance to several antimalarial drugs has been one of the major hindrances in control of malaria. Thus, the World Health Organization recommended artemisinin-based combination therapy (ACT) as a front-line treatment for malaria. The emergence of parasites resistant to artemisinin, along with resistant to ACT partner drugs, has led to ACT treatment failure. The artemisinin resistance is mostly related to the mutations in the propeller domain of the kelch13 (k13) gene that encodes protein Kelch13 (K13). The K13 protein has an important role in parasite reaction to oxidative stress. The most widely spread mutation in K13, with the highest degree of resistance, is a C580Y mutation. Other mutations, which are already identified as markers of artemisinin resistance, are R539T, I543T, and Y493H. The objective of this review is to provide current molecular insights into artemisinin resistance in Plasmodium falciparum. The trending use of artemisinin beyond its antimalarial effect is described. Immediate challenges and future research directions are discussed. Better understanding of the molecular mechanisms underlying artemisinin resistance will accelerate implementation of scientific findings to solve problems with malarial infection.
Collapse
Affiliation(s)
- Wihda Aisarul Azmi
- Eijkman Research Center for Molecular Biology, National Research and Innovation Agency, Cibinong, Bogor 16911, Indonesia; Master's Programme in Biomedical Sciences, Faculty of Medicine Universitas Indonesia, Jakarta 10430, Indonesia
| | - Andita Fitri Mutiara Rizki
- Eijkman Research Center for Molecular Biology, National Research and Innovation Agency, Cibinong, Bogor 16911, Indonesia; Master's Programme in Biomedical Sciences, Faculty of Medicine Universitas Indonesia, Jakarta 10430, Indonesia
| | - Yenny Djuardi
- Department of Parasitology, Faculty of Medicine Universitas Indonesia, Jakarta 10430, Indonesia
| | - I Made Artika
- Eijkman Research Center for Molecular Biology, National Research and Innovation Agency, Cibinong, Bogor 16911, Indonesia; Department of Biochemistry, Faculty of Mathematics and Natural Sciences, Bogor Agricultural University, Bogor 16680, Indonesia
| | - Josephine Elizabeth Siregar
- Eijkman Research Center for Molecular Biology, National Research and Innovation Agency, Cibinong, Bogor 16911, Indonesia.
| |
Collapse
|
6
|
Mariano RMDS, Gonçalves AAM, de Oliveira DS, Ribeiro HS, Pereira DFS, Santos IS, Lair DF, da Silva AV, Galdino AS, Chávez-Fumagalli MA, da Silveira-Lemos D, Dutra WO, Giunchetti RC. A Review of Major Patents on Potential Malaria Vaccine Targets. Pathogens 2023; 12:pathogens12020247. [PMID: 36839519 PMCID: PMC9959516 DOI: 10.3390/pathogens12020247] [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: 12/31/2022] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Malaria is a parasitic infection that is a great public health concern and is responsible for high mortality rates worldwide. Different strategies have been employed to improve disease control, demonstrating the ineffectiveness of controlling vectors, and parasite resistance to antimalarial drugs requires the development of an effective preventive vaccine. There are countless challenges to the development of such a vaccine directly related to the parasite's complex life cycle. After more than four decades of basic research and clinical trials, the World Health Organization (WHO) has recommended the pre-erythrocytic Plasmodium falciparum (RTS, S) malaria vaccine for widespread use among children living in malaria-endemic areas. However, there is a consensus that major improvements are needed to develop a vaccine with a greater epidemiological impact in endemic areas. This review discusses novel strategies for malaria vaccine design taking the target stages within the parasite cycle into account. The design of the multi-component vaccine shows considerable potential, especially as it involves transmission-blocking vaccines (TBVs) that eliminate the parasite's replication towards sporozoite stage parasites during a blood meal of female anopheline mosquitoes. Significant improvements have been made but additional efforts to achieve an efficient vaccine are required to improve control measures. Different strategies have been employed, thus demonstrating the ineffectiveness in controlling vectors, and parasite resistance to antimalarial drugs requires the development of a preventive vaccine. Despite having a vaccine in an advanced stage of development, such as the RTS, S malaria vaccine, the search for an effective vaccine against malaria is far from over. This review discusses novel strategies for malaria vaccine design taking into account the target stages within the parasite's life cycle.
Collapse
Affiliation(s)
- Reysla Maria da Silveira Mariano
- Laboratory of Biology of Cell Interactions, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte CEP 31270-901, MG, Brazil
| | - Ana Alice Maia Gonçalves
- Laboratory of Biology of Cell Interactions, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte CEP 31270-901, MG, Brazil
| | - Diana Souza de Oliveira
- Laboratory of Biology of Cell Interactions, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte CEP 31270-901, MG, Brazil
| | - Helen Silva Ribeiro
- Laboratory of Biology of Cell Interactions, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte CEP 31270-901, MG, Brazil
| | - Diogo Fonseca Soares Pereira
- Laboratory of Biology of Cell Interactions, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte CEP 31270-901, MG, Brazil
| | - Ingrid Soares Santos
- Laboratory of Biology of Cell Interactions, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte CEP 31270-901, MG, Brazil
| | - Daniel Ferreira Lair
- Laboratory of Biology of Cell Interactions, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte CEP 31270-901, MG, Brazil
| | - Augusto Ventura da Silva
- Laboratory of Biology of Cell Interactions, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte CEP 31270-901, MG, Brazil
| | - Alexsandro Sobreira Galdino
- Laboratory of Biotechnology of Microorganisms, Federal University of São João Del-Rei, Divinópolis CEP 35501-296, MG, Brazil
| | - Miguel Angel Chávez-Fumagalli
- Computational Biology and Chemistry Research Group, Vicerrectorado de Investigación, Universidad Católica de Santa María, Urb. San José S/N, Arequipa 04000, Peru
| | - Denise da Silveira-Lemos
- Campus Jaraguá, University José of Rosário Vellano, UNIFENAS, Belo Horizonte CEP 31270-901, MG, Brazil
| | - Walderez Ornelas Dutra
- Laboratory of Biology of Cell Interactions, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte CEP 31270-901, MG, Brazil
| | - Rodolfo Cordeiro Giunchetti
- Laboratory of Biology of Cell Interactions, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte CEP 31270-901, MG, Brazil
- Correspondence: or ; Tel.: +55-31-3409-3003
| |
Collapse
|
7
|
Wilks LR, Joshi G, Kang SM, Wang BZ, Gill HS. Peptide Cross-Linking Using Tyrosine Residues Facilitated by an Exogenous Nickel-Histidine Complex: A Facile Approach for Enhancing Vaccine-Specific Immunogenicity. ACS Infect Dis 2022; 8:2389-2395. [PMID: 36346898 DOI: 10.1021/acsinfecdis.2c00265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
An improved method for the generation of peptide vaccines using di-tyrosine cross-linking is described. The conserved ion channel peptide, M2e, of influenza A virus was modified with the addition of small tyrosine-rich regions (GYGY-) at both the N- and C-termini and extensively cross-linked via tyrosine-tyrosine linkages to form peptide nanoclusters. The cross-linking was catalyzed using exogenous nickel(II) ions complexed to an exogenous glycine-glycine-histidine peptide in the presence of an oxidizer. Mice that were intranasally or intramuscularly immunized with the M2e-vaccine nanoclusters induced comparable levels of M2e-specific serum antibodies. Vaccination via the intranasal or intramuscular route protected mice from subsequent lethal challenge with an influenza A virus. In comparison to our previous approach, where a histidine-rich tag was added into the peptide structure, the use of exogenous histidine reduced irrelevant off-target immune response. Additionally, the purity of the resulting nanoclusters is an attractive feature, making this approach appealing for vaccine development.
Collapse
Affiliation(s)
- Logan R Wilks
- Department of Chemical Engineering, Texas Tech University, 8th street and Canton Ave., Mail Stop 3121, Lubbock, Texas 79409-3121, United States
| | - Gaurav Joshi
- Department of Chemical Engineering, Texas Tech University, 8th street and Canton Ave., Mail Stop 3121, Lubbock, Texas 79409-3121, United States
| | - Sang-Moo Kang
- Institute for Biomedical Sciences, Georgia State University, 100 Piedmont Ave., Atlanta, Georgia 30302, United States
| | - Bao-Zhong Wang
- Institute for Biomedical Sciences, Georgia State University, 100 Piedmont Ave., Atlanta, Georgia 30302, United States
| | - Harvinder Singh Gill
- Department of Chemical Engineering, Texas Tech University, 8th street and Canton Ave., Mail Stop 3121, Lubbock, Texas 79409-3121, United States
| |
Collapse
|
8
|
Samy MN, Mahmoud BK, Shady NH, Abdelmohsen UR, Ross SA. Bioassay-Guided Fractionation with Antimalarial and Antimicrobial Activities of Paeonia officinalis. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27238382. [PMID: 36500473 PMCID: PMC9739537 DOI: 10.3390/molecules27238382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 11/24/2022] [Accepted: 11/25/2022] [Indexed: 12/03/2022]
Abstract
Bioassay-guided fractionation technique of roots of Paeonia officinalis led to isolation and structure elucidation of seven known compounds, including four monoterpene glycosides: lactiflorin (1), paeoniflorin (4), galloyl paeoniflorin (5), and (Z)-(1S,5R)-β-pinen-10-yl β-vicianoside (7); two phenolics: benzoic acid (2) and methyl gallate (3); and one sterol glycoside: β-sitosterol 3-O-β-D-glucopyranoside (6). The different fractions and the isolated compounds were evaluated for their antimicrobial and antimalarial activities. Fraction II and III showed antifungal activity against Candida neoformans with IC50 values of 28.11 and 74.37 µg/mL, respectively, compared with the standard fluconazole (IC50 = 4.68 µg/mL), and antibacterial potential against Pseudomonas aeruginosa (IC50 = 20.27 and 24.82 µg/mL, respectively) and Klebsiella pneumoniae (IC50 = 43.21 and 94.4 µg/mL, respectively), compared with the standard meropenem (IC50 = 28.67 and 43.94 µg/mL, respectively). Compounds 3 and 5 showed antimalarial activity against Plasmodium falciparum D6 with IC50 values of 1.57 and 4.72 µg/mL and P. falciparum W2 with IC50 values of 0.61 and 2.91 µg/mL, respectively, compared with the standard chloroquine (IC50 = 0.026 and 0.14 µg/mL, respectively).
Collapse
Affiliation(s)
- Mamdouh Nabil Samy
- Department of Pharmacognosy, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
- National Center for Natural Products Research, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA
| | - Basma Khalaf Mahmoud
- Department of Pharmacognosy, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
| | - Nourhan Hisham Shady
- Department of Pharmacognosy, Faculty of Pharmacy, Deraya University, New Minia 61111, Egypt
| | - Usama Ramadan Abdelmohsen
- Department of Pharmacognosy, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
- Department of Pharmacognosy, Faculty of Pharmacy, Deraya University, New Minia 61111, Egypt
- Correspondence: (U.R.A.); (S.A.R.)
| | - Samir Anis Ross
- National Center for Natural Products Research, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA
- Department of BioMolecular Science, Division of Phamacognosy, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA
- Correspondence: (U.R.A.); (S.A.R.)
| |
Collapse
|
9
|
Sun Y, Shi X, Lu F, Fu H, Yin Y, Xu J, Jin C, Han ET, Huang X, Chen Y, Dong C, Cheng Y. Vesicular stomatitis virus-based vaccine targeting plasmodium blood-stage antigens elicits immune response and protects against malaria with protein booster strategy. Front Microbiol 2022; 13:1042414. [PMID: 36504817 PMCID: PMC9731671 DOI: 10.3389/fmicb.2022.1042414] [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: 09/12/2022] [Accepted: 11/07/2022] [Indexed: 11/25/2022] Open
Abstract
Merozoite invasion of the erythrocytes in humans is a key step in the pathogenesis of malaria. The proteins involved in the merozoite invasion could be potential targets for the development of malaria vaccines. Novel viral-vector-based malaria vaccine regimens developed are currently under clinical trials. Vesicular stomatitis virus (VSV) is a single-stranded negative-strand RNA virus widely used as a vector for virus or cancer vaccines. Whether the VSV-based malarial vaccine is more effective than conventional vaccines based on proteins involved in parasitic invasion is still unclear. In this study, we have used the reverse genetics system to construct recombinant VSVs (rVSVs) expressing apical membrane protein 1 (AMA1), rhoptry neck protein 2 (RON2), and reticulocyte-binding protein homolog 5 (RH5), which are required for Plasmodium falciparum invasion. Our results showed that VSV-based viral vaccines significantly increased Plasmodium-specific IgG levels and lymphocyte proliferation. Also, VSV-PyAMA1 and VSV-PyRON2sp prime-boost regimens could significantly increase the levels of IL-2 and IFN-γ-producing by CD4+ and CD8+ T cells and suppress invasion in vitro. The rVSV prime-protein boost regimen significantly increase Plasmodium antigen-specific IgG levels in the serum of mice compared to the homologous rVSV prime-boost. Furthermore, the protective efficacy of rVSV prime protein boost immunization in the mice challenged with P. yoelii 17XL was better compared to traditional antigen immunization. Together, our results show that VSV vector is a novel strategy for malarial vaccine development and preventing the parasitic diseases.
Collapse
Affiliation(s)
- Yifan Sun
- Department of Laboratory Medicine, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China,Laboratory of Pathogen Infection and Immunity, Department of Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Xiaodan Shi
- Laboratory of Pathogen Infection and Immunity, Department of Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Feng Lu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Haitian Fu
- Laboratory of Pathogen Infection and Immunity, Department of Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China,Department of Nuclear Medicine, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Yi Yin
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Jiahui Xu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Cheng Jin
- Department of Hepatobiliary Surgery, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Eun-taek Han
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, South Korea
| | - Xuan Huang
- Department of Laboratory Medicine, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Yongquan Chen
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Chunsheng Dong
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu, China,*Correspondence: Chunsheng Dong,
| | - Yang Cheng
- Laboratory of Pathogen Infection and Immunity, Department of Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China,Yang Cheng,
| |
Collapse
|
10
|
Frimpong A, Ofori MF, Degoot AM, Kusi KA, Gershom B, Quartey J, Kyei-Baafour E, Nguyen N, Ndifon W. Perturbations in the T cell receptor β repertoire during malaria infection in children: A preliminary study. Front Immunol 2022; 13:971392. [PMID: 36311775 PMCID: PMC9606469 DOI: 10.3389/fimmu.2022.971392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 09/22/2022] [Indexed: 11/13/2022] Open
Abstract
The changes occurring in the T cell repertoire during clinical malaria infection in children remain unknown. In this study, we undertook the first detailed comparative study of the T cell repertoire in African children with and without clinical malaria to test the hypothesis that clonotypic expansions that occur during P. falciparum infection will contribute to the generation of a T cell repertoire that is unique to each disease state. We profiled the complementarity-determining region 3 (CDR3) of the TCRβ chain sequences from children with Plasmodium falciparum infections (asymptomatic, uncomplicated and severe malaria) and compared these with sequences from healthy children. Interestingly, we discovered that children with symptomatic malaria have a lower TCR diversity and frequency of shared (or “public”) TCR sequences compared to asymptomatic children. Also, TCR diversity was inversely associated with parasitemia. Furthermore, by clustering TCR sequences based on their predicted antigen specificities, we identified a specificity cluster, with a 4-mer amino acid motif, that is overrepresented in the asymptomatic group compared to the diseased groups. Further investigations into this finding may help in delineating important antigenic targets for vaccine and therapeutic development. The results show that the T cell repertoire in children is altered during malaria, suggesting that exposure to P. falciparum antigens disrupts the adaptive immune response, which is an underlying feature of the disease.
Collapse
Affiliation(s)
- Augustina Frimpong
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), Department of Biochemistry, Cell, and Molecular Biology, University of Ghana, Accra, Ghana
- Immunology Department, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
- African Institute for Mathematical Sciences, Accra, Ghana
- *Correspondence: Wilfred Ndifon, ; Augustina Frimpong,
| | - Michael Fokuo Ofori
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), Department of Biochemistry, Cell, and Molecular Biology, University of Ghana, Accra, Ghana
- Immunology Department, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Abdoelnaser M. Degoot
- Research Department, African Institute for Mathematical Sciences, Next Einstein Initiative, Kigali, Rwanda
| | - Kwadwo Asamoah Kusi
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), Department of Biochemistry, Cell, and Molecular Biology, University of Ghana, Accra, Ghana
- Immunology Department, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Buri Gershom
- African Institute for Mathematical Sciences, Cape Town, South Africa
| | - Jacob Quartey
- Immunology Department, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Eric Kyei-Baafour
- Immunology Department, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
| | | | - Wilfred Ndifon
- Research Department, African Institute for Mathematical Sciences, Next Einstein Initiative, Kigali, Rwanda
- African Institute for Mathematical Sciences, Cape Town, South Africa
- *Correspondence: Wilfred Ndifon, ; Augustina Frimpong,
| |
Collapse
|
11
|
Hassanzadeh P, Atyabi F, Dinarvand R. Nanobionics: From plant empowering to the infectious disease treatment. J Control Release 2022; 349:890-901. [PMID: 35901860 DOI: 10.1016/j.jconrel.2022.07.028] [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: 05/11/2022] [Revised: 07/20/2022] [Accepted: 07/21/2022] [Indexed: 10/16/2022]
Abstract
Infectious diseases (ID) are serious threats against the global health and socio-economic conditions. Vaccination usually plays a key role in disease prevention, however, insufficient efficiency or immunogenicity may be quite challenging. Using the advanced vectors for delivery of vaccines with suitable efficiency, safety, and immune-modulatory activity, and tunable characteristics could be helpful, but there are no systematic reviews confirming the capabilities of the vaccine delivery systems for covering various types of pathogens. Furthermore, high rates of the infections, transmission, and fatal ratio and diversity of the pathogens and infection mechanisms may negatively influence vaccine effectiveness. The absence of highly-effective antibiotics against the resistant strains of bacteria and longevity of antibiotic testing have provoked increasing needs towards the application of more accurate and specific theranostic strategies including the nanotechnology-based ones. Nanobionics which is based on the charge storage and transport in the molecular structures, could be of key value in the molecular diagnostic tests and highly-specific electro-analytical methods or devices. Such devices based on the early disease diagnostics might be of critical significance against various types of diseases. This article highlights the significance of nanobionics against ID.
Collapse
Affiliation(s)
- Parichehr Hassanzadeh
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 13169-43551, Iran; Sasan Hospital, Tehran 14159-83391, Iran.
| | - Fatemeh Atyabi
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 13169-43551, Iran
| | - Rassoul Dinarvand
- Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 13169-43551, Iran
| |
Collapse
|
12
|
Zhang J, Li J, Hu K, Zhou Q, Chen X, He J, Yin S, Chi Y, Liao X, Xiao Y, Qin H, Zheng Z, Chen J. Screening Novel Vaccine Candidates for Leishmania Donovani by Combining Differential Proteomics and Immunoinformatics Analysis. Front Immunol 2022; 13:902066. [PMID: 35812381 PMCID: PMC9260594 DOI: 10.3389/fimmu.2022.902066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 05/25/2022] [Indexed: 11/18/2022] Open
Abstract
Visceral leishmaniasis (VL), also known as kala-azar, is the most dangerous form of leishmaniasis. Currently no effective vaccine is available for clinical use. Since the pathogenicity of different Leishmania strains is inconsistent, the differentially expressed proteins in Leishmania strains may play an important role as virulence factors in pathogenesis. Therefore, effective vaccine candidate targets may exist in the differentially expressed proteins. In this study, we used differential proteomics analysis to find the differentially expressed proteins in two Leishmania donovani strains, and combined with immunoinformatics analysis to find new vaccine candidates. The differentially expressed proteins from L. DD8 (low virulent) and L. 9044 (virulent) strains were analyzed by LC-MS/MS, and preliminarily screened by antigenicity, allergenicity and homology evaluation. The binding peptides of MHC II, IFN-γ and MHC I from differentially expressed proteins were then predicted and calculated for the second screening. IFN-γ/IL-10 ratios and conserved domain prediction were performed to choose more desirable differentially expressed proteins. Finally, the 3D structures of three vaccine candidate proteins were produced and submitted for molecular dynamics simulation and molecular docking interaction with TLR4/MD2. The results showed that 396 differentially expressed proteins were identified by LC-MS/MS, and 155 differentially expressed proteins were selected through antigenicity, allergenicity and homology evaluation. Finally, 16 proteins whose percentages of MHC II, IFN-γ and MHC I binding peptides were greater than those of control groups (TSA, LmSTI1, LeIF, Leish-111f) were considered to be suitable vaccine candidates. Among the 16 candidates, amino acid permease, amastin-like protein and the hypothetical protein (XP_003865405.1) simultaneously had the large ratios of IFN-γ/IL-10 and high percentages of MHC II, IFN-γ and MHC I, which should be focused on. In conclusion, our comprehensive work provided a methodological basis to screen new vaccine candidates for a better intervention against VL and associated diseases.
Collapse
Affiliation(s)
- Jianhui Zhang
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Jiao Li
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Kaifeng Hu
- Department of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qi Zhou
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Xiaoxiao Chen
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Jinlei He
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Shuangshuang Yin
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Yangjian Chi
- Department of Urinary Surgery, Jianou Municipal Hospital of Fujian Province, Jianou, China
| | - Xuechun Liao
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Yuying Xiao
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Hanxiao Qin
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| | - Zhiwan Zheng
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
- *Correspondence: Zhiwan Zheng, ; Jianping Chen,
| | - Jianping Chen
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
- Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Sichuan University, Chengdu, China
- *Correspondence: Zhiwan Zheng, ; Jianping Chen,
| |
Collapse
|
13
|
Stochastic expression of invasion genes in Plasmodium falciparum schizonts. Nat Commun 2022; 13:3004. [PMID: 35637187 PMCID: PMC9151791 DOI: 10.1038/s41467-022-30605-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 04/22/2022] [Indexed: 12/15/2022] Open
Abstract
Genetically identical cells are known to exhibit differential phenotypes in the same environmental conditions. These phenotypic variants are linked to transcriptional stochasticity and have been shown to contribute towards adaptive flexibility of a wide range of unicellular organisms. Here, we investigate transcriptional heterogeneity and stochastic gene expression in Plasmodium falciparum by performing the quasilinear multiple annealing and looping based amplification cycles (MALBAC) based amplification and single cell RNA sequencing of blood stage schizonts. Our data reveals significant transcriptional variations in the schizont stage with a distinct group of highly variable invasion gene transcripts being identified. Moreover, the data reflects several diversification processes including putative developmental “checkpoint”; transcriptomically distinct parasite sub-populations and transcriptional switches in variable gene families (var, rifin, phist). Most of these features of transcriptional variability are preserved in isogenic parasite cell populations (albeit with a lesser amplitude) suggesting a role of epigenetic factors in cell-to-cell transcriptional variations in human malaria parasites. Lastly, we apply quantitative RT-PCR and RNA-FISH approach and confirm stochastic expression of key invasion genes, such as, msp1, msp3, msp7, eba181 and ama1 which represent prime candidates for invasion-blocking vaccines. Genetically identical cells can be phenotypically diverse to allow adaptive flexibility in a given environment. This phenotypic diversity is driven by epigenetic and transcriptional variability. Here, Tripathi et al. perform scRNA-seq of isogenic and non-isogenic Plasmodium falciparum schizont populations to explore transcriptional heterogeneity and stochastic gene expression during the course of development.
Collapse
|
14
|
Siddiqui AJ, Bhardwaj J, Hamadou WS, Goyal M, Jahan S, Ashraf SA, Jamal A, Sharma P, Sachidanandan M, Badraoui R, Snoussi M, Adnan M. Impact of chemoprophylaxis immunisation under halofantrine (CPS-HF) drug cover in Plasmodium yoelii Swiss mice malaria model. Folia Parasitol (Praha) 2022; 69. [PMID: 35145048 DOI: 10.14411/fp.2022.003] [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: 06/30/2021] [Accepted: 11/28/2021] [Indexed: 11/19/2022]
Abstract
In the present study, we have investigated the role of antimalarial drug halofantrine (HF) in inducing the sterile protection against challenges with sporozoites of the live infectious Plasmodium yoelii (Killick-Kendrick, 1967) in Swiss mice malaria model. We observed that during the first to third sequential sporozoite inoculation cycles, blood-stage patency remains the same in the control and chemoprophylaxis under HF drug cover (CPS-HF) groups. However, a delayed blood-stage infection was observed during the fourth and fifth sporozoite challenges and complete sterile protection was produced following the sixth sporozoite challenge in CPS-HF mice. We also noticed a steady decline in liver stage parasite load after 3th to 6th sporozoite challenge cycle in CPS-HF mice. CPS-HF immunisation results in a significant up-regulation of pro-inflammatory cytokines (IFN-γ, TNF-α, IL-12 and iNOS) and down-regulation of anti-inflammatory cytokines (IL-10 and TGF-β) mRNA expression in hepatic mononuclear cells (HMNC) and spleen cells in the immunised CPS-HF mice (after 6th sporozoite challenge) compared to control. Overall, our study suggests that the repetitive sporozoite inoculation under HF drug treatment develops a strong immune response that confers protection against subsequent challenges with sporozoites of P. yoelii.
Collapse
Affiliation(s)
- Arif Jamal Siddiqui
- Department of Biology, College of Science, University of Hail, Hail, Saudi Arabia.,Molecular Parasitology and Immunology Division, CSIR-Central Drug Research Institute, Lucknow, India.,Both authors have contributed equally to this work and share first authorship.,Corresponding author
| | - Jyoti Bhardwaj
- Indiana University, School of Medicine, Indianapolis, Indiana, United States.,Molecular Parasitology and Immunology Division, CSIR-Central Drug Research Institute, Lucknow, India.,Both authors have contributed equally to this work and share first authorship
| | - Walid Sabri Hamadou
- Department of Biology, College of Science, University of Hail, Hail, Saudi Arabia
| | - Manish Goyal
- Molecular Parasitology and Immunology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Sadaf Jahan
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al Majmaah, Saudi Arabia
| | - Syed Amir Ashraf
- Department of Clinical Nutrition, College of Applied Medial Sciences, University of Hail, Hail, Saudi Arabia
| | - Arshad Jamal
- Department of Biology, College of Science, University of Hail, Hail, Saudi Arabia
| | - Pankaj Sharma
- Molecular Parasitology and Immunology Division, CSIR-Central Drug Research Institute, Lucknow, India.,Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, U.S.A
| | | | - Riadh Badraoui
- Department of Biology, College of Science, University of Hail, Hail, Saudi Arabia
| | - Mejdi Snoussi
- Department of Biology, College of Science, University of Hail, Hail, Saudi Arabia
| | - Mohd Adnan
- Department of Biology, College of Science, University of Hail, Hail, Saudi Arabia
| |
Collapse
|
15
|
Labis da Costa MJ, Nascimento GC, Athie TS, Sales Silva JD, Reis EA, Martin AP, Godman B, Dias Godói IP. Willingness to pay for a hypothetical malaria vaccine in Brazil: a cross-sectional study and the implications. J Comp Eff Res 2022; 11:263-274. [PMID: 35029122 DOI: 10.2217/cer-2021-0073] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: Malaria is an infection caused by protozoa of genus Plasmodium, considered the one associated with increasingly large outbreaks. Methods: A cross-sectional study was conducted with residents in the northern region of Brazil on the willingness to pay (WTP) for a hypothetical vaccine against malaria (effective protection of 80%). Results: Of 616 people interviewed, most interviewees were female (61%) and were employed (97%). The median individual maximum WTP for a hypothetical malaria vaccine was US$11.90 (BRL 50). Conclusion: The northern region of Brazil is one of the largest markets for a malaria vaccine due to its epidemiological relevance. Consequently, economic studies will be important to assist in the assessment of the potential price and value of new vaccines.
Collapse
Affiliation(s)
- Maria José Labis da Costa
- Instute of Health & Biological Studies - Universidade Federal do Sul e Sudeste do Pará, Avenida dos Ipês, s/n, Cidade Universitária, Cidade Jardim, Marabá, Pará 68.500-000, Brazil
| | - Gesiane Cavalcante Nascimento
- Instute of Health & Biological Studies - Universidade Federal do Sul e Sudeste do Pará, Avenida dos Ipês, s/n, Cidade Universitária, Cidade Jardim, Marabá, Pará 68.500-000, Brazil
| | - Thannuse Silva Athie
- Instute of Health & Biological Studies - Universidade Federal do Sul e Sudeste do Pará, Avenida dos Ipês, s/n, Cidade Universitária, Cidade Jardim, Marabá, Pará 68.500-000, Brazil
| | - Juliana de Sales Silva
- Institute of Studies in Agricultural & Regional Development - Universidade Federal do Sul e Sudeste do Pará, Avenida dos Ipês, s/n, Cidade Universitária, Cidade Jardim, Marabá, Pará 68.500-000, Brazil
| | - Edna Afonso Reis
- Departament of Stascs, Exact Sciences Institute, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil.,Researcher of the Group (CNPq) for Epidemiological, Economic & Pharmacological Studies of Arboviruses (EEPIFARBO) - Universidade Federal do Sul e Sudeste do Pará; Avenida dos Ipês, s/n, Cidade Universitária, Cidade Jardim, Marabá, Pará, Brazil
| | - Antony Paul Martin
- Faculty of Health & Life Sciences, Brownlow Hill, Liverpool L69 3BX, UK.,QC Medica, York, North Yorkshire, UK
| | - Brian Godman
- Department of Pharmacoepidemiology, Institute of Pharmacy & Biomedical Sciences, 161 Cathedral Street, Glasgow G4 0RE, UK.,Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Isabella Piassi Dias Godói
- Instute of Health & Biological Studies - Universidade Federal do Sul e Sudeste do Pará, Avenida dos Ipês, s/n, Cidade Universitária, Cidade Jardim, Marabá, Pará 68.500-000, Brazil.,Researcher of the Group (CNPq) for Epidemiological, Economic & Pharmacological Studies of Arboviruses (EEPIFARBO) - Universidade Federal do Sul e Sudeste do Pará; Avenida dos Ipês, s/n, Cidade Universitária, Cidade Jardim, Marabá, Pará, Brazil
| |
Collapse
|
16
|
Review of the Current Landscape of the Potential of Nanotechnology for Future Malaria Diagnosis, Treatment, and Vaccination Strategies. Pharmaceutics 2021; 13:pharmaceutics13122189. [PMID: 34959470 PMCID: PMC8706932 DOI: 10.3390/pharmaceutics13122189] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 12/13/2021] [Accepted: 12/14/2021] [Indexed: 12/24/2022] Open
Abstract
Malaria eradication has for decades been on the global health agenda, but the causative agents of the disease, several species of the protist parasite Plasmodium, have evolved mechanisms to evade vaccine-induced immunity and to rapidly acquire resistance against all drugs entering clinical use. Because classical antimalarial approaches have consistently failed, new strategies must be explored. One of these is nanomedicine, the application of manipulation and fabrication technology in the range of molecular dimensions between 1 and 100 nm, to the development of new medical solutions. Here we review the current state of the art in malaria diagnosis, prevention, and therapy and how nanotechnology is already having an incipient impact in improving them. In the second half of this review, the next generation of antimalarial drugs currently in the clinical pipeline is presented, with a definition of these drugs' target product profiles and an assessment of the potential role of nanotechnology in their development. Opinions extracted from interviews with experts in the fields of nanomedicine, clinical malaria, and the economic landscape of the disease are included to offer a wider scope of the current requirements to win the fight against malaria and of how nanoscience can contribute to achieve them.
Collapse
|
17
|
Alum Pickering Emulsion as Effective Adjuvant to Improve Malaria Vaccine Efficacy. Vaccines (Basel) 2021; 9:vaccines9111244. [PMID: 34835175 PMCID: PMC8624716 DOI: 10.3390/vaccines9111244] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 09/27/2021] [Accepted: 10/15/2021] [Indexed: 12/12/2022] Open
Abstract
Malaria is a life-threatening global epidemic disease and has caused more than 400,000 deaths in 2019. To control and prevent malaria, the development of a vaccine is a potential method. An effective malaria vaccine should either combine antigens from all stages of the malaria parasite’s life cycle, or epitopes of multiple key antigens due to the complexity of the Plasmodium parasite. Malaria’s random constructed antigen-1 (M.RCAg-1) is one of the recombinant vaccines, which was selected from a DNA library containing thousands of diverse multi-epitope chimeric antigen genes. Moreover, besides selecting an antigen, using an adjuvant is another important procedure for most vaccine development procedures. Freund’s adjuvant is considered an effective vaccine adjuvant for malaria vaccine, but it cannot be used in clinical settings because of its serious side effects. Traditional adjuvants, such as alum adjuvant, are limited by their unsatisfactory immune effects in malaria vaccines, hence there is an urgent need to develop a novel, safe and efficient adjuvant. In recent years, Pickering emulsions have attracted increasing attention as novel adjuvant. In contrast to classical emulsions, Pickering emulsions are stabilized by solid particles instead of surfactant, having pliability and lateral mobility. In this study, we selected aluminum hydroxide gel (termed as “alum”) as a stabilizer to prepare alum-stabilized Pickering emulsions (ALPE) as a malaria vaccine adjuvant. In addition, monophosphoryl lipid A (MPLA) as an immunostimulant was incorporated into the Pickering emulsion (ALMPE) to further enhance the immune response. In vitro tests showed that, compared with alum, ALPE and ALMPE showed higher antigen load rates and could be effectively endocytosed by J774a.1 cells. In vivo studies indicated that ALMPE could induce as high antibody titers as Freund’s adjuvant. The biocompatibility study also proved ALMPE with excellent biocompatibility. These results suggest that ALMPE is a potential adjuvant for a malaria vaccine.
Collapse
|
18
|
Patra S, Singh M, Wasnik K, Pareek D, Gupta PS, Mukherjee S, Paik P. Polymeric Nanoparticle Based Diagnosis and Nanomedicine for Treatment and Development of Vaccines for Cerebral Malaria: A Review on Recent Advancement. ACS APPLIED BIO MATERIALS 2021; 4:7342-7365. [PMID: 35006689 DOI: 10.1021/acsabm.1c00635] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cerebral malaria occurs due to Plasmodium falciparum infection, which causes 228 million infections and 450,000 deaths worldwide every year. African people are mostly affected with nearly 91% cases, of which 86% are pregnant women and infants. India and Brazil are the other two countries severely suffering from malaria endemicity. Commonly used drugs have severe side effects, and unfortunately no suitable vaccine is available in the market today. In this line, this review is focused on polymeric nanomaterials and nanocapsules that can be used for the development of effective diagnostic strategies, nanomedicines, and vaccines in the management of cerebral malaria. Further, this review will help scientists and medical professionals by updating the status on the development stages of polymeric nanoparticle based diagnostics, nanomedicines, and vaccines and strategies to eradicate cerebral malaria. In addition to this, the predominant focus of this review is antimalarial agents based on polymer nanomedicines that are currently in the preclinical and clinical trial stages, and potential developments are suggested as well. This review further will have an important social and commercial impact worldwide for the development of polymeric nanomedicines and strategies for the treatment of cerebral malaria.
Collapse
Affiliation(s)
- Sukanya Patra
- School of Biomedical Engineering, Indian Institute of Technology-BHU, Varanasi 221005, India
| | - Monika Singh
- School of Biomedical Engineering, Indian Institute of Technology-BHU, Varanasi 221005, India
| | - Kirti Wasnik
- School of Biomedical Engineering, Indian Institute of Technology-BHU, Varanasi 221005, India
| | - Divya Pareek
- School of Biomedical Engineering, Indian Institute of Technology-BHU, Varanasi 221005, India
| | - Prem Shankar Gupta
- School of Biomedical Engineering, Indian Institute of Technology-BHU, Varanasi 221005, India
| | - Sudip Mukherjee
- Department of Bioengineering, Rice University, Houston, Texas 77030, United States
| | - Pradip Paik
- School of Biomedical Engineering, Indian Institute of Technology-BHU, Varanasi 221005, India
| |
Collapse
|
19
|
Wilks LR, Joshi G, Grisham MR, Gill HS. Tyrosine-Based Cross-Linking of Peptide Antigens to Generate Nanoclusters with Enhanced Immunogenicity: Demonstration Using the Conserved M2e Peptide of Influenza A. ACS Infect Dis 2021; 7:2723-2735. [PMID: 34432416 DOI: 10.1021/acsinfecdis.1c00219] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
A method of creating nanoclusters (NCs) from soluble peptide molecules is described utilizing an approach based on a tyrosine-tyrosine cross-linking reaction. A reactive tag comprising histidine and tyrosine residues was introduced at the termini of the peptide molecules. The cross-linking reaction led to the creation of dityrosine bonds within the tag, which allowed for the generation of peptide NCs. We show that it is essential for the reactive tag to be present at both the "N" and "C" termini of the peptide for cluster formation to occur. Additionally, the cross-linking reaction was systematically characterized to show the importance of reaction conditions on final cluster diameter, allowing us to generate NCs of various sizes. To demonstrate the immunogenic potential of the peptide clusters, we chose to study the conserved influenza peptide, M2e, as the antigen. M2e NCs were formulated using the cross-linking reaction. We show the ability of the clusters to generate protective immunity in a dose, size, and frequency dependent manner against a lethal influenza A challenge in BALB/c mice. Taken together, the data presented suggest this new cluster formation technique can generate highly immunogenic peptide NCs in a simple and controllable manner.
Collapse
Affiliation(s)
- Logan R. Wilks
- Department of Chemical Engineering, Texas Tech University, 8th Street and Canton Avenue, Mail Stop 3121, Lubbock, Texas 79409-3121, United States
| | - Gaurav Joshi
- Department of Chemical Engineering, Texas Tech University, 8th Street and Canton Avenue, Mail Stop 3121, Lubbock, Texas 79409-3121, United States
| | - Megan R. Grisham
- Department of Chemical Engineering, Texas Tech University, 8th Street and Canton Avenue, Mail Stop 3121, Lubbock, Texas 79409-3121, United States
| | - Harvinder Singh Gill
- Department of Chemical Engineering, Texas Tech University, 8th Street and Canton Avenue, Mail Stop 3121, Lubbock, Texas 79409-3121, United States
| |
Collapse
|
20
|
Pan C, Yue H, Zhu L, Ma GH, Wang HL. Prophylactic vaccine delivery systems against epidemic infectious diseases. Adv Drug Deliv Rev 2021; 176:113867. [PMID: 34280513 PMCID: PMC8285224 DOI: 10.1016/j.addr.2021.113867] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/20/2021] [Accepted: 07/11/2021] [Indexed: 01/04/2023]
Abstract
Prophylactic vaccines have evolved from traditional whole-cell vaccines to safer subunit vaccines. However, subunit vaccines still face problems, such as poor immunogenicity and low efficiency, while traditional adjuvants are usually unable to meet specific response needs. Advanced delivery vectors are important to overcome these barriers; they have favorable safety and effectiveness, tunable properties, precise location, and immunomodulatory capabilities. Nevertheless, there has been no systematic summary of the delivery systems to cover a wide range of infectious pathogens. We herein summarized and compared the delivery systems for major or epidemic infectious diseases caused by bacteria, viruses, fungi, and parasites. We also included the newly licensed vaccines (e.g., COVID-19 vaccines) and those close to licensure. Furthermore, we highlighted advanced delivery systems with high efficiency, cross-protection, or long-term protection against epidemic pathogens, and we put forward prospects and thoughts on the development of future prophylactic vaccines.
Collapse
Affiliation(s)
- Chao Pan
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing 100071, PR China
| | - Hua Yue
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Li Zhu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing 100071, PR China
| | - Guang-Hui Ma
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China.
| | - Heng-Liang Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing 100071, PR China.
| |
Collapse
|
21
|
Guiding the Immune Response to a Conserved Epitope in MSP2, an Intrinsically Disordered Malaria Vaccine Candidate. Vaccines (Basel) 2021; 9:vaccines9080855. [PMID: 34451980 PMCID: PMC8402609 DOI: 10.3390/vaccines9080855] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/16/2021] [Accepted: 07/29/2021] [Indexed: 02/06/2023] Open
Abstract
The malaria vaccine candidate merozoite surface protein 2 (MSP2) has shown promise in clinical trials and is in part responsible for a reduction in parasite densities. However, strain-specific reductions in parasitaemia suggested that polymorphic regions of MSP2 are immuno-dominant. One strategy to bypass the hurdle of strain-specificity is to bias the immune response towards the conserved regions. Two mouse monoclonal antibodies, 4D11 and 9H4, recognise the conserved C-terminal region of MSP2. Although they bind overlapping epitopes, 4D11 reacts more strongly with native MSP2, suggesting that its epitope is more accessible on the parasite surface. In this study, a structure-based vaccine design approach was applied to the intrinsically disordered antigen, MSP2, using a crystal structure of 4D11 Fv in complex with its minimal binding epitope. Molecular dynamics simulations and surface plasmon resonance informed the design of a series of constrained peptides that mimicked the 4D11-bound epitope structure. These peptides were conjugated to keyhole limpet hemocyanin and used to immunise mice, with high to moderate antibody titres being generated in all groups. The specificities of antibody responses revealed that a single point mutation can focus the antibody response towards a more favourable epitope. This structure-based approach to peptide vaccine design may be useful not only for MSP2-based malaria vaccines, but also for other intrinsically disordered antigens.
Collapse
|
22
|
Rachmania S, Sulistyaningsih E, Ratna Dewi AAI. Recombinant DBL2β-PfEMP1 of the Indonesian Plasmodium falciparum induces immune responses in Wistar rats. J Taibah Univ Med Sci 2021; 16:422-430. [PMID: 34140870 PMCID: PMC8178645 DOI: 10.1016/j.jtumed.2020.12.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 12/12/2020] [Accepted: 12/17/2020] [Indexed: 11/21/2022] Open
Abstract
OBJECTIVES The Duffy binding-like (DBL) domain of the Plasmodium falciparum Erythrocyte Membrane Protein 1 (PfEMP1) is reportedly responsible for the pathophysiology of cerebral malaria. People living in endemic malaria areas possess specific antibodies against PfEMP1 and elicit immune responses to control the severity of malaria infection. Therefore, PfEMP1 may be a potential protein-based vaccine candidate. This study aimed to explore the humoral and cellular immune responses induced by the recombinant DBL2β-PfEMP1 obtained from the Indonesian P. falciparum isolate. METHODS The recombinant protein was expressed in Escherichia coli BL21(DE3) as soluble and insoluble fractions, and this protein was purified using affinity chromatography before administration as a subcutaneous injection in Wistar rats on days 1, 21, and 42. Sera were harvested 14 days after the second and third injections to determine the titre of IgG and the concentration of CD4+ cells using the enzyme linked immunosorbent assay (ELISA). RESULTS The IgG titre and the CD4+ cell concentration were found to be increased after the second and third injections. The Mann-Whitney test results showed a significant difference between the control and treatment groups for both the IgG and CD4+ cells (p = 0.001 and p = 0.000, respectively). Western blotting results indicated the presence of a specific antibody against the recombinant DBL2β-PfEMP1. CONCLUSIONS The recombinant DBL2β-PfEMP1 of the Indonesian P. falciparum isolate could induce humoral and cellular immune responses. Further studies on IgG exerting inhibitory effects and the role of CD4+ cells and their association with other effector cells are essential to determine the efficacy of DBL2β-PfEMP1 and its potential application as a peptide-based malaria vaccine candidate.
Collapse
Affiliation(s)
- Sheilla Rachmania
- Department of Histology, Faculty of Medicine, University of Jember, Indonesia
| | - Erma Sulistyaningsih
- Department of Parasitology, Faculty of Medicine, University of Jember, Indonesia
| | - Anak Agung I. Ratna Dewi
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, University of Jember, Indonesia
| |
Collapse
|
23
|
Chuang GY, Shen CH, Cheung CSF, Gorman J, Creanga A, Joyce MG, Leung K, Rawi R, Wang L, Yang ES, Yang Y, Zhang B, Zhang Y, Kanekiyo M, Zhou T, DeKosky BJ, Graham BS, Mascola JR, Kwong PD. Sequence-Signature Optimization Enables Improved Identification of Human HV6-1-Derived Class Antibodies That Neutralize Diverse Influenza A Viruses. Front Immunol 2021; 12:662909. [PMID: 34135892 PMCID: PMC8201785 DOI: 10.3389/fimmu.2021.662909] [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: 02/01/2021] [Accepted: 04/26/2021] [Indexed: 11/13/2022] Open
Abstract
Sequence signatures of multidonor broadly neutralizing influenza antibodies can be used to quantify the prevalence of B cells with virus-neutralizing potential to accelerate development of broadly protective vaccine strategies. Antibodies of the same class share similar recognition modes and developmental pathways, and several antibody classes have been identified that neutralize diverse group 1- and group 2-influenza A viruses and have been observed in multiple human donors. One such multidonor antibody class, the HV6-1-derived class, targets the stem region of hemagglutinin with extraordinary neutralization breadth. Here, we use an iterative process to combine informatics, biochemical, and structural analyses to delineate an improved sequence signature for HV6-1-class antibodies. Based on sequence and structure analyses of known HV6-1 class antibodies, we derived a more inclusive signature (version 1), which we used to search for matching B-cell transcripts from published next-generation sequencing datasets of influenza vaccination studies. We expressed selected antibodies, evaluated their function, and identified amino acid-level requirements from which to refine the sequence signature (version 2). The cryo-electron microscopy structure for one of the signature-identified antibodies in complex with hemagglutinin confirmed motif recognition to be similar to known HV6-1-class members, MEDI8852 and 56.a.09, despite differences in recognition-loop length. Threading indicated the refined signature to have increased accuracy, and signature-identified heavy chains, when paired with the light chain of MEDI8852, showed neutralization comparable to the most potent members of the class. Incorporating sequences of additional class members thus enables an improved sequence signature for HV6-1-class antibodies, which can identify class members with increased accuracy.
Collapse
Affiliation(s)
- Gwo-Yu Chuang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Chen-Hsiang Shen
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Crystal Sao-Fong Cheung
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Jason Gorman
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Adrian Creanga
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - M Gordon Joyce
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Kwanyee Leung
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Reda Rawi
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Lingshu Wang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Eun Sung Yang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Yongping Yang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Baoshan Zhang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Yi Zhang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Masaru Kanekiyo
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Tongqing Zhou
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Brandon J DeKosky
- Department of Pharmaceutical Chemistry and Department of Chemical Engineering, University of Kansas, Lawrence, KS, United States
| | - Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - John R Mascola
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Peter D Kwong
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
24
|
Bamgbose T, Anvikar AR, Alberdi P, Abdullahi IO, Inabo HI, Bello M, Cabezas-Cruz A, de la Fuente J. Functional Food for the Stimulation of the Immune System Against Malaria. Probiotics Antimicrob Proteins 2021; 13:1254-1266. [PMID: 33791994 PMCID: PMC8012070 DOI: 10.1007/s12602-021-09780-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/23/2021] [Indexed: 12/20/2022]
Abstract
Drug resistance has become a threat to global health, and new interventions are needed to control major infectious diseases. The composition of gut microbiota has been linked to human health and has been associated with severity of malaria. Fermented foods contribute to the community of healthy gut bacteria. Despite the studies connecting gut microbiota to the prevention of malaria transmission and severity, research on developing functional foods for the purpose of manipulating the gut microbiota for malaria control is limited. This review summarizes recent knowledge on the role of the gut microbiota in malaria prevention and treatment. This information should encourage the search for lactic acid bacteria expressing α-Gal and those that exhibit the desired immune stimulating properties for the development of functional food and probiotics for malaria control.
Collapse
Affiliation(s)
- Timothy Bamgbose
- ICMR, -National Institute of Malaria Research, Sector 8, Dwarka, New Delhi, India
- Department of Microbiology, Ahmadu Bello University, Samaru Zaria, Kaduna, Nigeria
| | - Anupkumar R Anvikar
- ICMR, -National Institute of Malaria Research, Sector 8, Dwarka, New Delhi, India
| | - Pilar Alberdi
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo s/n, 13005, Ciudad Real, Spain
| | - Isa O Abdullahi
- Department of Microbiology, Ahmadu Bello University, Samaru Zaria, Kaduna, Nigeria
| | - Helen I Inabo
- Department of Microbiology, Ahmadu Bello University, Samaru Zaria, Kaduna, Nigeria
| | - Mohammed Bello
- Department of Veterinary Public Health and Preventive Medicine, Ahmadu Bello University, Samaru Zaria, Kaduna, Nigeria
| | - Alejandro Cabezas-Cruz
- UMR BIPAR, INRAE, ANSES, Ecole Nationale Vétérinaire D'Alfort, Université Paris-Est, 94700, Maisons-Alfort, France
| | - José de la Fuente
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo s/n, 13005, Ciudad Real, Spain.
- Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK, 74078, USA.
| |
Collapse
|
25
|
Amiah MA, Ouattara A, Okou DT, N'Guetta SPA, Yavo W. Polymorphisms in Fc Gamma Receptors and Susceptibility to Malaria in an Endemic Population. Front Immunol 2020; 11:561142. [PMID: 33281811 PMCID: PMC7689034 DOI: 10.3389/fimmu.2020.561142] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 10/05/2020] [Indexed: 11/13/2022] Open
Abstract
Repeated infections by Plasmodium falciparum result in a humoral response that could reduce disease symptoms and prevent the development of clinical malaria. The principal mechanism underlying this humoral response is that immunoglobulin G (IgG) binds directly to the parasites, thus causing their neutralization. However, the action of antibodies alone is not always sufficient to eliminate pathogens from an organism. One key element involved in the recognition of IgG that plays a crucial role in the destruction of the parasites responsible for spreading malaria is the family of Fc gamma receptors. These receptors are expressed on the surface of immune cells. Several polymorphisms have been detected in the genes encoding these receptors, associated with susceptibility or resistance to malaria in different populations. In this review, we describe identified polymorphisms within the family of Fc gamma receptors and the impact of these variations on the response of a host to infection as well as provide new perspectives for the design of an effective vaccine for malaria.
Collapse
Affiliation(s)
- Mireille Ahou Amiah
- Malaria Research and Control Center, National Public Health Institute, Abidjan, Côte d'Ivoire.,Laboratory of Genetics, Unité de Formation et de Recherche (UFR) BIOSCIENCES, Félix Houphouët-Boigny University, Abidjan, Côte d'Ivoire
| | - Amed Ouattara
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - David Tea Okou
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Simon-Pierre Assanvo N'Guetta
- Laboratory of Genetics, Unité de Formation et de Recherche (UFR) BIOSCIENCES, Félix Houphouët-Boigny University, Abidjan, Côte d'Ivoire
| | - William Yavo
- Malaria Research and Control Center, National Public Health Institute, Abidjan, Côte d'Ivoire.,Department of Parasitology and Mycology, Faculty of Pharmacy, Félix Houphouët-Boigny University, Abidjan, Côte d'Ivoire
| |
Collapse
|
26
|
Roth G, Gale EC, Alcántara-Hernández M, Luo W, Axpe E, Verma R, Yin Q, Yu AC, Lopez Hernandez H, Maikawa CL, Smith AAA, Davis MM, Pulendran B, Idoyaga J, Appel EA. Injectable Hydrogels for Sustained Codelivery of Subunit Vaccines Enhance Humoral Immunity. ACS CENTRAL SCIENCE 2020; 6:1800-1812. [PMID: 33145416 PMCID: PMC7596866 DOI: 10.1021/acscentsci.0c00732] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Indexed: 05/15/2023]
Abstract
Vaccines aim to elicit a robust, yet targeted, immune response. Failure of a vaccine to elicit such a response arises in part from inappropriate temporal control over antigen and adjuvant presentation to the immune system. In this work, we sought to exploit the immune system's natural response to extended pathogen exposure during infection by designing an easily administered slow-delivery vaccine platform. We utilized an injectable and self-healing polymer-nanoparticle (PNP) hydrogel platform to prolong the codelivery of vaccine components to the immune system. We demonstrated that these hydrogels exhibit unique delivery characteristics, whereby physicochemically distinct compounds (such as antigen and adjuvant) could be codelivered over the course of weeks. When administered in mice, hydrogel-based sustained vaccine exposure enhanced the magnitude, duration, and quality of the humoral immune response compared to standard PBS bolus administration of the same model vaccine. We report that the creation of a local inflammatory niche within the hydrogel, coupled with sustained exposure of vaccine cargo, enhanced the magnitude and duration of germinal center responses in the lymph nodes. This strengthened germinal center response promoted greater antibody affinity maturation, resulting in a more than 1000-fold increase in antigen-specific antibody affinity in comparison to bolus immunization. In summary, this work introduces a simple and effective vaccine delivery platform that increases the potency and durability of subunit vaccines.
Collapse
Affiliation(s)
- Gillie
A. Roth
- Department
of Bioengineering, Stanford University, Stanford, California 94305, United States
| | - Emily C. Gale
- Department
of Biochemistry, Stanford University School
of Medicine, Stanford, California 94305, United States
| | - Marcela Alcántara-Hernández
- Department
of Microbiology & Immunology, Stanford
University School of Medicine, Stanford, California 94305, United States
- Program
in Immunology, Stanford University School
of Medicine, Stanford, California 94305, United States
| | - Wei Luo
- Institute
for Immunity, Transplantation & Infection, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Eneko Axpe
- Department
of Materials Science & Engineering, Stanford University, Stanford, California 94305, United States
| | - Rohit Verma
- Institute
for Immunity, Transplantation & Infection, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Qian Yin
- Institute
for Immunity, Transplantation & Infection, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Anthony C. Yu
- Department
of Materials Science & Engineering, Stanford University, Stanford, California 94305, United States
| | - Hector Lopez Hernandez
- Department
of Materials Science & Engineering, Stanford University, Stanford, California 94305, United States
| | - Caitlin L. Maikawa
- Department
of Bioengineering, Stanford University, Stanford, California 94305, United States
| | - Anton A. A. Smith
- Department
of Materials Science & Engineering, Stanford University, Stanford, California 94305, United States
| | - Mark M. Davis
- Department
of Microbiology & Immunology, Stanford
University School of Medicine, Stanford, California 94305, United States
- Program
in Immunology, Stanford University School
of Medicine, Stanford, California 94305, United States
- Institute
for Immunity, Transplantation & Infection, Stanford University School of Medicine, Stanford, California 94305, United States
- The
Howard Hughes Medical Institute, Stanford
University School of Medicine, Stanford, California 94305, United States
| | - Bali Pulendran
- Department
of Microbiology & Immunology, Stanford
University School of Medicine, Stanford, California 94305, United States
- Program
in Immunology, Stanford University School
of Medicine, Stanford, California 94305, United States
- Institute
for Immunity, Transplantation & Infection, Stanford University School of Medicine, Stanford, California 94305, United States
- Department
of Pathology, Stanford University School
of Medicine, Stanford, California 94305, United States
- ChEM-H
Institute, Stanford University, Stanford, California 94305, United States
| | - Juliana Idoyaga
- Department
of Microbiology & Immunology, Stanford
University School of Medicine, Stanford, California 94305, United States
- Program
in Immunology, Stanford University School
of Medicine, Stanford, California 94305, United States
- Institute
for Immunity, Transplantation & Infection, Stanford University School of Medicine, Stanford, California 94305, United States
- ChEM-H
Institute, Stanford University, Stanford, California 94305, United States
| | - Eric A. Appel
- Department
of Bioengineering, Stanford University, Stanford, California 94305, United States
- Institute
for Immunity, Transplantation & Infection, Stanford University School of Medicine, Stanford, California 94305, United States
- Department
of Materials Science & Engineering, Stanford University, Stanford, California 94305, United States
- ChEM-H
Institute, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
27
|
Su XZ, Zhang C, Joy DA. Host-Malaria Parasite Interactions and Impacts on Mutual Evolution. Front Cell Infect Microbiol 2020; 10:587933. [PMID: 33194831 PMCID: PMC7652737 DOI: 10.3389/fcimb.2020.587933] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 09/22/2020] [Indexed: 12/22/2022] Open
Abstract
Malaria is the most deadly parasitic disease, affecting hundreds of millions of people worldwide. Malaria parasites have been associated with their hosts for millions of years. During the long history of host-parasite co-evolution, both parasites and hosts have applied pressure on each other through complex host-parasite molecular interactions. Whereas the hosts activate various immune mechanisms to remove parasites during an infection, the parasites attempt to evade host immunity by diversifying their genome and switching expression of targets of the host immune system. Human intervention to control the disease such as antimalarial drugs and vaccination can greatly alter parasite population dynamics and evolution, particularly the massive applications of antimalarial drugs in recent human history. Vaccination is likely the best method to prevent the disease; however, a partially protective vaccine may have unwanted consequences that require further investigation. Studies of host-parasite interactions and co-evolution will provide important information for designing safe and effective vaccines and for preventing drug resistance. In this essay, we will discuss some interesting molecules involved in host-parasite interactions, including important parasite antigens. We also discuss subjects relevant to drug and vaccine development and some approaches for studying host-parasite interactions.
Collapse
Affiliation(s)
- Xin-Zhuan Su
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Cui Zhang
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Deirdre A Joy
- Parasitology and International Programs Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
28
|
Nateghi Rostami M. CRISPR/Cas9 gene drive technology to control transmission of vector‐borne parasitic infections. Parasite Immunol 2020; 42:e12762. [DOI: 10.1111/pim.12762] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 05/20/2020] [Accepted: 05/26/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Mahmoud Nateghi Rostami
- Laboratory of Biology of Host‐Parasite Interactions Department of Parasitology Pasteur Institute of Iran Tehran Iran
| |
Collapse
|
29
|
Imai T, Suzue K, Ngo-Thanh H, Shimokawa C, Hisaeda H. Potential and Limitations of Cross-Protective Vaccine against Malaria by Blood-Stage Naturally Attenuated Parasite. Vaccines (Basel) 2020; 8:vaccines8030375. [PMID: 32664476 PMCID: PMC7564742 DOI: 10.3390/vaccines8030375] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/07/2020] [Accepted: 07/09/2020] [Indexed: 01/13/2023] Open
Abstract
Human malaria vaccine trials have revealed vaccine efficacy but improvement is still needed. In this study, we aimed to re-evaluate vaccination with blood-stage naturally attenuated parasites, as a whole-organism vaccine model against cross-strain and cross-species malaria, to establish a better vaccination strategy. C57BL/6 mice controlled blood-stage Plasmodium yoelii 17XNL (PyNL) within 1 month of infection, while mice with a variety of immunodeficiencies demonstrated different susceptibilities to PyNL, including succumbing to hyperparasitemia. However, after recovery, survivors had complete protection against a challenge with the lethal strain PyL. Unlike cross-strain protection, PyNL-recovered mice failed to induce sterile immunity against Plasmodium berghei ANKA, although prolonged survival was observed in some vaccinated mice. Splenomegaly is a typical characteristic of malaria; the splenic structure became reorganized to prioritize extra-medullary hematopoiesis and to eliminate parasites. We also found that the peritoneal lymph node was enlarged, containing activated/memory phenotype cells that did not confer protection against PyL challenge. Hemozoins remained in the spleen several months after PyNL infection. Generation of an attenuated human blood-stage parasite expressing proteins from multiple species of malaria would greatly improve anti-malaria vaccination.
Collapse
Affiliation(s)
- Takashi Imai
- Department of Infectious Diseases and Host Defense, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan; (K.S.); (H.N.-T.)
- Department of Parasitology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
- Correspondence: ; Tel.: +81-27-220-8023
| | - Kazutomo Suzue
- Department of Infectious Diseases and Host Defense, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan; (K.S.); (H.N.-T.)
| | - Ha Ngo-Thanh
- Department of Infectious Diseases and Host Defense, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan; (K.S.); (H.N.-T.)
| | - Chikako Shimokawa
- Department of Parasitology, National Institute of Infectious Diseases, Tokyo 162-0052, Japan; (C.S.); (H.H.)
| | - Hajime Hisaeda
- Department of Parasitology, National Institute of Infectious Diseases, Tokyo 162-0052, Japan; (C.S.); (H.H.)
| |
Collapse
|
30
|
The use of proteomics for the identification of promising vaccine and diagnostic biomarkers in Plasmodium falciparum. Parasitology 2020; 147:1255-1262. [PMID: 32618524 DOI: 10.1017/s003118202000102x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Plasmodium falciparum is the main cause of severe malaria in humans that can lead to death. There is growing evidence of drug-resistance in P. falciparum treatment, and the design of effective vaccines remains an ongoing strategy to control the disease. On the other hand, the recognition of specific diagnostic markers for P. falciparum can accelerate the diagnosis of this parasite in the early stages of infection. Therefore, the identification of novel antigenic proteins especially by proteomic tools is urgent for vaccination and diagnosis of P. falciparum. The proteome diversity of the life cycle stages of P. falciparum, the altered proteome of P. falciparum-infected human sera and altered proteins in P. falciparum-infected erythrocytes could be proposed as appropriate proteins for the aforementioned aims. Accordingly, this review highlights and proposes different proteins identified using proteomic approaches as promising markers in the diagnosis and vaccination of P. falciparum. It seems that most of the candidates identified in this study were able to elicit immune responses in the P. falciparum-infected hosts and they also played major roles in the life cycle, pathogenicity and key pathways of this parasite.
Collapse
|
31
|
Geiger KM, Guignard D, Yang C, Bikorimana JP, Correia BE, Houard S, Mkindi C, Daubenberger CA, Spertini F, Corradin G, Audran R. Epitope Mapping and Fine Specificity of Human T and B Cell Responses for Novel Candidate Blood-Stage Malaria Vaccine P27A. Front Immunol 2020; 11:412. [PMID: 32210975 PMCID: PMC7076177 DOI: 10.3389/fimmu.2020.00412] [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: 12/03/2019] [Accepted: 02/21/2020] [Indexed: 11/13/2022] Open
Abstract
P27A is a novel synthetic malaria vaccine candidate derived from the blood stage Plasmodium falciparum protein Trophozoite Exported Protein 1 (TEX1/PFF0165c). In phase 1a/1b clinical trials in malaria unexposed adults in Switzerland and in malaria pre-exposed adults in Tanzania, P27A formulated with Alhydrogel and GLA-SE adjuvants induced antigen-specific antibodies and T-cell activity. The GLA-SE adjuvant induced significantly stronger humoral responses than the Alhydrogel adjuvant. Groups of pre-exposed and unexposed subjects received identical vaccine formulations, which supported the comparison of the cellular and humoral response to P27A in terms of fine specificity and affinity for populations and adjuvants. Globally, fine specificity of the T and B cell responses exhibited preferred recognized sequences and did not highlight major differences between adjuvants or populations. Affinity of anti-P27A antibodies was around 10-8 M in all groups. Pre-exposed volunteers presented anti-P27A with higher affinity than unexposed volunteers. Increasing the dose of GLA-SE from 2.5 to 5 μg in pre-exposed volunteers improved anti-P27A affinity and decreased the number of recognized epitopes. These results indicate a higher maturation of the humoral response in pre-exposed volunteers, particularly when immunized with P27A formulated with 5 μg GLA-SE.
Collapse
Affiliation(s)
- Kristina M Geiger
- Biochemistry Department, University of Lausanne, Epalinges, Switzerland.,Department of Infectious Diseases and Immunity, University of California, Berkeley, Berkeley, CA, United States
| | - Daniel Guignard
- Biochemistry Department, University of Lausanne, Epalinges, Switzerland
| | - Che Yang
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.,Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Jean-Pierre Bikorimana
- Division of Immunology and Allergy, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Bruno E Correia
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.,Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | | | - Catherine Mkindi
- Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland.,Department of Intervention and Clinical Trials, Ifakara Health Institute, Bagamoyo, Tanzania
| | - Claudia A Daubenberger
- Swiss Tropical and Public Health Institute, Basel, Switzerland.,University of Basel, Basel, Switzerland
| | - François Spertini
- Division of Immunology and Allergy, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | | | - Régine Audran
- Division of Immunology and Allergy, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| |
Collapse
|
32
|
Bettencourt P. Current Challenges in the Identification of Pre-Erythrocytic Malaria Vaccine Candidate Antigens. Front Immunol 2020; 11:190. [PMID: 32153565 PMCID: PMC7046804 DOI: 10.3389/fimmu.2020.00190] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 01/24/2020] [Indexed: 12/27/2022] Open
Abstract
Plasmodium spp.-infected mosquitos inject sporozoites into the skin of a mammalian host during a blood meal. These enter the host's circulatory system and establish an infection in the liver. After a silent metamorphosis, merozoites invade the blood leading to the symptomatic and transmissible stages of malaria. The silent pre-erythrocytic malaria stage represents a bottleneck in the disease which is ideal to block progression to clinical malaria, through chemotherapeutic and immunoprophylactic interventions. RTS,S/AS01, the only malaria vaccine close to licensure, although with poor efficacy, blocks the sporozoite invasion mainly through the action of antibodies against the CSP protein, a major component of the pellicle of the sporozoite. Strikingly, sterile protection against malaria can be obtained through immunization with radiation-attenuated sporozoites, genetically attenuated sporozoites or through chemoprophylaxis with infectious sporozoites in animals and humans, but the deployability of sporozoite-based live vaccines pose tremendous challenges. The protection induced by sporozoites occurs in the pre-erythrocytic stages and is mediated mainly by antibodies against the sporozoite and CD8+ T cells against peptides presented by MHC class I molecules in infected hepatocytes. Thus, the identification of malaria antigens expressed in the sporozoite and liver-stage may provide new vaccine candidates to be included, alone or in combination, as recombinant protein-based, virus-like particles or sub-unit virally-vectored vaccines. Here I review the efforts being made to identify Plasmodium falciparum antigens expressed during liver-stage with focus on the development of parasite, hepatocyte, mouse models, and resulting rate of infection in order to identify new vaccine candidates and to improve the efficacy of the current vaccines. Finally, I propose new approaches for the identification of liver-stage antigens based on immunopeptidomics.
Collapse
|
33
|
Quadiri A, Kalia I, Kashif M, Singh AP. Identification and characterization of protective CD8 + T-epitopes in a malaria vaccine candidate SLTRiP. IMMUNITY INFLAMMATION AND DISEASE 2020; 8:50-61. [PMID: 31967737 PMCID: PMC7016849 DOI: 10.1002/iid3.283] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 12/20/2019] [Accepted: 12/23/2019] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Efforts are required at developing an effective vaccine that can inhibit malaria prevalence and transmission. Identifying the critical immunogenic antigens and understanding their interactions with host proteins forms a major focus of subunit vaccine development. Previously, our laboratory showed that SLTRiP conferred protection to the liver stage of Plasmodium growth in rodents. In the follow-up of earlier research, we demonstrate that SLTRiP-mediated protection is majorly concentrated in specific regions of protein. METHOD To identify particular protective regions of protein, we synthesized multiple nonoverlapping fragments from SLTRiP protein. From this, we designed a panel of 8-20mer synthetic peptides, which were predicted using T-epitope-based prediction algorithm. We utilized the IFN-γ enzyme-linked immunosorbent spot assay to identify immunodominant peptides. The latter were used to immunize mice, and these mice were challenged to assess protection. RESULTS The protective polypeptide fragment SLTRiP C3 and SLTRiP C4 were identified, by expressing and testing multiple fragments of PbSLTRiP protein. The immune responses generated by these fragments were compared to identify the immunodominant fragment. The T-epitopes were predicted from SLTRiP protein using computer-based algorithms. The in vitro immune responses generated by these peptides were compared with each other to identify the immunodominant T-epitope. Immunization using these peptides showed significant reduction in parasite numbers during liver stage. CONCLUSION Our findings show that the protective efficacy shown by SLTRiP is localized in particular protein fragments. The peptides designed from such regions showed protective efficacy equivalent to whole protein. The sequence conservation analysis with human Plasmodium species also showed that these peptides were conserved. In conclusion, these peptides or their equivalent from other Plasmodium species could impart protection against malaria in their respective hosts too. Our studies provide a basis for the inclusion of these peptides in clinical vaccine constructs against malaria.
Collapse
Affiliation(s)
- Afshana Quadiri
- Infectious Diseases Laboratory, National Institute of Immunology, New Delhi, India
| | - Inderjeet Kalia
- Infectious Diseases Laboratory, National Institute of Immunology, New Delhi, India
| | - Mohammad Kashif
- Infectious Diseases Laboratory, National Institute of Immunology, New Delhi, India
| | - Agam P Singh
- Infectious Diseases Laboratory, National Institute of Immunology, New Delhi, India
| |
Collapse
|
34
|
Loiseau C, Cooper MM, Doolan DL. Deciphering host immunity to malaria using systems immunology. Immunol Rev 2019; 293:115-143. [PMID: 31608461 DOI: 10.1111/imr.12814] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 09/18/2019] [Accepted: 09/20/2019] [Indexed: 12/11/2022]
Abstract
A century of conceptual and technological advances in infectious disease research has changed the face of medicine. However, there remains a lack of effective interventions and a poor understanding of host immunity to the most significant and complex pathogens, including malaria. The development of successful interventions against such intractable diseases requires a comprehensive understanding of host-pathogen immune responses. A major advance of the past decade has been a paradigm switch in thinking from the contemporary reductionist (gene-by-gene or protein-by-protein) view to a more holistic (whole organism) view. Also, a recognition that host-pathogen immunity is composed of complex, dynamic interactions of cellular and molecular components and networks that cannot be represented by any individual component in isolation. Systems immunology integrates the field of immunology with omics technologies and computational sciences to comprehensively interrogate the immune response at a systems level. Herein, we describe the system immunology toolkit and report recent studies deploying systems-level approaches in the context of natural exposure to malaria or controlled human malaria infection. We contribute our perspective on the potential of systems immunity for the rational design and development of effective interventions to improve global public health.
Collapse
Affiliation(s)
- Claire Loiseau
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Qld, Australia
| | - Martha M Cooper
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Qld, Australia
| | - Denise L Doolan
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Qld, Australia
| |
Collapse
|