1
|
Yadav M, Dahiya N, Srivastava V, Singh H, Kataria D, Janjoter S, Dixit R, Sehrawat N. Molecular characterization, expression and in-silico analysis of fibrinogen-related protein 1 (frep1) in malaria vector Anopheles stephensi. Mol Biol Rep 2024; 51:970. [PMID: 39249121 DOI: 10.1007/s11033-024-09891-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 08/26/2024] [Indexed: 09/10/2024]
Abstract
BACKGROUND Fibrinogen-related protein 1 (frep1) is a member of the pattern-recognizing receptor family (PRR) which generates an innate immune response after recognizing the pattern associated molecular pattern (PAMP) that occurs on the surface of microorganisms. The main objective of this study is to characterize frep1 and its in-silico analysis in Anopheles stephensi. METHODS AND RESULT The DNA was extracted from female Anopheles stephensi. PCR was performed for complete analysis of frep1 using specific primers. The gene sequence of frep1 was identified by Sanger sequencing. The bioinformatics structure analysis approach revealed the presence of 3 exons and 4 introns in the frep1. The sequence of frep1 was submitted to NCBI GeneBank with accession number ON817187.1. Quantitative real-time PCR was performed to analyze frep1 expression. At the developmental stage, frep1 is highly expressed in the L1 stage, egg, and adult female mosquito. In addition, frep1 is highly expressed in the tissue fat body, midgut, and salivary gland. After blood-fed, an upregulation of frep1 at 48 h in the midgut, and downregulation in fat body were observed at different time intervals. CONCLUSION The genomic data of frep1 is encoded by 12,443 bp. The frep1 has a significant role in the early metamorphosis. Its expression in fat body and midgut suggests it could be important for fat metabolism and post-blood digestion. The conserved domain could be targeted for vector control. Further study is required to elucidate its function against malaria parasites to confirm its agonist role in malaria transmission.
Collapse
Affiliation(s)
- Mahima Yadav
- Department of Genetics, Maharshi Dayanand University, Rohtak, Haryana, 124001, India
| | - Nisha Dahiya
- Department of Genetics, Maharshi Dayanand University, Rohtak, Haryana, 124001, India
| | - Vartika Srivastava
- National Institute of Malaria Research (NIMR), Sector 8, Dwarka, New Delhi, 110077, India
| | - Hitesh Singh
- Department of Genetics, Maharshi Dayanand University, Rohtak, Haryana, 124001, India
| | - Divya Kataria
- Department of Genetics, Maharshi Dayanand University, Rohtak, Haryana, 124001, India
| | - Sangeeta Janjoter
- Department of Genetics, Maharshi Dayanand University, Rohtak, Haryana, 124001, India
| | - Ranjnikant Dixit
- National Institute of Malaria Research (NIMR), Sector 8, Dwarka, New Delhi, 110077, India
| | - Neelam Sehrawat
- Department of Genetics, Maharshi Dayanand University, Rohtak, Haryana, 124001, India.
| |
Collapse
|
2
|
Wang J, Chen K, Yang J, Zhang S, Li Y, Liu G, Luo J, Yin H, Wang G, Guan G. Comparative genomic analysis of Babesia duncani responsible for human babesiosis. BMC Biol 2022; 20:153. [PMID: 35790982 PMCID: PMC9258201 DOI: 10.1186/s12915-022-01361-9] [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: 02/22/2022] [Accepted: 06/23/2022] [Indexed: 11/29/2022] Open
Abstract
Background Human babesiosis, caused by parasites of the genus Babesia, is an emerging and re-emerging tick-borne disease that is mainly transmitted by tick bites and infected blood transfusion. Babesia duncani has caused majority of human babesiosis in Canada; however, limited data are available to correlate its genomic information and biological features. Results We generated a B. duncani reference genome using Oxford Nanopore Technology (ONT) and Illumina sequencing technology and uncovered its biological features and phylogenetic relationship with other Apicomplexa parasites. Phylogenetic analyses revealed that B. duncani form a clade distinct from B. microti, Babesia spp. infective to bovine and ovine species, and Theileria spp. infective to bovines. We identified the largest species-specific gene family that could be applied as diagnostic markers for this pathogen. In addition, two gene families show signals of significant expansion and several genes that present signatures of positive selection in B. duncani, suggesting their possible roles in the capability of this parasite to infect humans or tick vectors. Conclusions Using ONT sequencing and Illumina sequencing technologies, we provide the first B. duncani reference genome and confirm that B. duncani forms a phylogenetically distinct clade from other Piroplasm parasites. Comparative genomic analyses show that two gene families are significantly expanded in B. duncani and may play important roles in host cell invasion and virulence of B. duncani. Our study provides basic information for further exploring B. duncani features, such as host-parasite and tick-parasite interactions. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-022-01361-9.
Collapse
Affiliation(s)
- Jinming Wang
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Lanzhou, 730046, Gansu, China.
| | - Kai Chen
- Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Jifei Yang
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Lanzhou, 730046, Gansu, China
| | - Shangdi Zhang
- Department of Clinical Laboratory, The Second Hospital of Lanzhou University, Lanzhou, 730030, China
| | - Youquan Li
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Lanzhou, 730046, Gansu, China
| | - Guangyuan Liu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Lanzhou, 730046, Gansu, China
| | - Jianxun Luo
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Lanzhou, 730046, Gansu, China
| | - Hong Yin
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Lanzhou, 730046, Gansu, China.,Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou University, Yangzhou, 225009, China
| | - Guangying Wang
- Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China.
| | - Guiquan Guan
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Science, Lanzhou, 730046, Gansu, China.
| |
Collapse
|
3
|
Yu S, Wang J, Luo X, Zheng H, Wang L, Yang X, Wang Y. Transmission-Blocking Strategies Against Malaria Parasites During Their Mosquito Stages. Front Cell Infect Microbiol 2022; 12:820650. [PMID: 35252033 PMCID: PMC8889032 DOI: 10.3389/fcimb.2022.820650] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 01/31/2022] [Indexed: 11/24/2022] Open
Abstract
Malaria is still the most widespread parasitic disease and causes the most infections globally. Owing to improvements in sanitary conditions and various intervention measures, including the use of antimalarial drugs, the malaria epidemic in many regions of the world has improved significantly in the past 10 years. However, people living in certain underdeveloped areas are still under threat. Even in some well-controlled areas, the decline in malaria infection rates has stagnated or the rates have rebounded because of the emergence and spread of drug-resistant malaria parasites. Thus, new malaria control methods must be developed. As the spread of the Plasmodium parasite is dependent on the part of its life cycle that occurs in mosquitoes, to eliminate the possibility of malaria infections, transmission-blocking strategies against the mosquito stage should be the first choice. In fact, after the gametocyte enters the mosquito body, it undergoes a series of transformation processes over a short period, thus providing numerous potential blocking targets. Many research groups have carried out studies based on targeting the blocking of transmission during the mosquito phase and have achieved excellent results. Meanwhile, the direct killing of mosquitoes could also significantly reduce the probability of malaria infections. Microorganisms that display complex interactions with Plasmodium, such as Wolbachia and gut flora, have shown observable transmission-blocking potential. These could be used as a biological control strategy and play an important part in blocking the transmission of malaria.
Collapse
Affiliation(s)
- Shasha Yu
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing, China
| | - Jing Wang
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing, China
| | - Xue Luo
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing, China
| | - Hong Zheng
- Department of Thoracic Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Luhan Wang
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing, China
| | - Xuesen Yang
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing, China
| | - Ying Wang
- Department of Tropical Medicine, College of Military Preventive Medicine, Army Medical University, Chongqing, China
- *Correspondence: Ying Wang,
| |
Collapse
|
4
|
Viswanath VK, Gore ST, Valiyaparambil A, Mukherjee S, Lakshminarasimhan A. Plasmodium chitinases: revisiting a target of transmission-blockade against malaria. Protein Sci 2021; 30:1493-1501. [PMID: 33934433 DOI: 10.1002/pro.4095] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/28/2021] [Accepted: 04/30/2021] [Indexed: 01/21/2023]
Abstract
Malaria is a life-threatening disease caused by one of the five species of Plasmodium, among which Plasmodium falciparum cause the deadliest form of the disease. Plasmodium species are dependent on a vertebrate host and a blood-sucking insect vector to complete their life cycle. Plasmodium chitinases belonging to the GH18 family are secreted inside the mosquito midgut, during the ookinete stage of the parasite. Chitinases mediate the penetration of parasite through the peritrophic membrane, facilitating access to the gut epithelial layer. In this review, we describe Plasmodium chitinases with special emphasis on chitinases from P. falciparum and P. vivax, the representative examples of the short and long forms of this protein. In addition to the chitinase domain, chitinases belonging to the long form contain a pro-domain and chitin-binding domain. Amino acid sequence alignment of long and short form chitinase domains reveals multiple positions containing variant residues. A subset of these positions was found to be conserved or invariant within long or short forms, indicating the role of these positions in attributing form-specific activity. The reported differences in affinities to allosamidin for P. vivax and P. falciparum were predicted to be due to different residues at two amino acid positions, resulting in altered interactions with the inhibitor. Understanding the role of these amino acids in Plasmodium chitinases will help us elucidate the mechanism of catalysis and the mode of inhibition, which will be the key for identification of potent inhibitors or antibodies demonstrating transmission-blocking activity.
Collapse
Affiliation(s)
- Vysakh K Viswanath
- Tata Institute for Genetics and Society, Center at inStem, Bengaluru, India
| | - Suraj T Gore
- Aurigene Discovery Technologies Ltd, Bengaluru, India
| | | | | | | |
Collapse
|
5
|
Alzan HF, Bastos RG, Ueti MW, Laughery JM, Rathinasamy VA, Cooke BM, Suarez CE. Assessment of Babesia bovis 6cys A and 6cys B as components of transmission blocking vaccines for babesiosis. Parasit Vectors 2021; 14:210. [PMID: 33879245 PMCID: PMC8056569 DOI: 10.1186/s13071-021-04712-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 03/31/2021] [Indexed: 12/20/2022] Open
Abstract
Background Babesia bovis reproduces sexually in the gut of its tick vector Rhipicephalus microplus, which involves expression of 6cys A and 6cys B proteins. Members of the widely conserved 6cys superfamily are candidates for transmission blocking vaccines (TBV), but intricacies in the immunogenicity of the 6cys proteins in the related Plasmodium parasites required the identification of transmission blocking domains in these molecules for vaccine design. Hereby, the immunogenic efficacy of recombinant (r) B. bovis 6cys A and B proteins as a TBV formulation was studied. Methods The immunogenicity of r6cys A and 6cys B proteins expressed in a eukaryotic system was evaluated in a cattle immunization trial (3 immunized and 3 control calves). A B. bovis sexual stage induction in vitro inhibition assay to assess the ability of antibodies to block the production of sexual forms by the parasite was developed. Results Immunized cattle generated antibodies against r6cys A and r6cys B that were unable to block sexual reproduction of the parasite in ticks. Additionally, these antibodies also failed in recognizing native 6cys A and 6cys B and peptides representing 6cys A and 6cys B functional domains and in inhibiting the development of sexual forms in an in vitro induction system. In contrast, rabbit antibodies generated against synthetic peptides representing predicted B-cell epitopes of 6cys A and 6cys B recognized recombinant and native forms of both 6cys proteins as well as peptides representing 6cys A and 6cys B functional domains and were able to neutralize development of sexual forms of the parasite in vitro. Conclusions These data, combined with similar work performed on Plasmodium 6cys proteins, indicate that an effective 6cys protein-based TBV against B. bovis will require identifying and targeting selected regions of proteins containing epitopes able to reduce transmission. Graphic abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s13071-021-04712-7.
Collapse
Affiliation(s)
- Heba F Alzan
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, USA. .,Parasitology and Animal Diseases Department, National Research Center, Dokki, Giza, Egypt. .,Tick and Tick-Borne Disease Research Unit, National Research Center, Dokki, Giza, 12622, Egypt.
| | - Reginaldo G Bastos
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Massaro W Ueti
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, USA.,Animal Disease Research Unit, United States Department of Agricultural - Agricultural Research Service, Pullman, WA, USA
| | - Jacob M Laughery
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Vignesh A Rathinasamy
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - Brian M Cooke
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland, Australia
| | - Carlos E Suarez
- Department of Veterinary Microbiology and Pathology, College of Veterinary Medicine, Washington State University, Pullman, WA, USA. .,Animal Disease Research Unit, United States Department of Agricultural - Agricultural Research Service, Pullman, WA, USA.
| |
Collapse
|
6
|
Li D, Yu C, Guo J, Wang Y, Zhao Y, Wang L, Soe MT, Feng H, Kyaw MP, Sattabongkot J, Jiang L, Cui L, Zhu X, Cao Y. Plasmodium vivax HAP2/GCS1 gene exhibits limited genetic diversity among parasite isolates from the Greater Mekong Subregion. Parasit Vectors 2020; 13:175. [PMID: 32264948 PMCID: PMC7137254 DOI: 10.1186/s13071-020-04050-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 03/28/2020] [Indexed: 12/02/2022] Open
Abstract
Background Antigens expressed in sexual stages of the malaria parasites are targets of transmission-blocking vaccines (TBVs). HAP2/GCS1, a TBV candidate, is critical for fertilization in Plasmodium. Here, the genetic diversity of PvHAP2 was studied in Plasmodium vivax parasite populations from the Greater Mekong Subregion (GMS). Methods Plasmodium vivax clinical isolates were collected in clinics from the China-Myanmar border region (135 samples), western Thailand (41 samples) and western Myanmar (51 samples). Near full-length Pvhap2 (nucleotides 13–2574) was amplified and sequenced from these isolates. Molecular evolution studies were conducted to evaluate the genetic diversity, selection and population differentiation. Results Sequencing of the pvhap2 gene for a total of 227 samples from the three P. vivax populations revealed limited genetic diversity of this gene in the GMS (π = 0.00036 ± 0.00003), with the highest π value observed in Myanmar (0.00053 ± 0.00009). Y133S was the dominant mutation in the China-Myanmar border (99.26%), Myanmar (100%) and Thailand (95.12%). Results of all neutrality tests were negative for all the three populations, suggesting the possible action of purifying selection. Codon-based tests identified specific codons which are under purifying or positive selections. Wright’s fixation index showed low to moderate genetic differentiation of P. vivax populations in the GMS, with FST ranging from 0.04077 to 0.24833, whereas high levels of genetic differentiation were detected between the China-Myanmar border and Iran populations (FST = 0.60266), and between Thailand and Iran populations (FST = 0.44161). A total of 20 haplotypes were identified, with H2 being the abundant haplotype in China-Myanmar border, Myanmar and Thailand populations. Epitope mapping prediction of Pvhap2 antigen showed that high-score B-cell epitopes are located in the S307-G324, L429-P453 and V623-D637 regions. The E317K and D637N mutations located within S307-G324 and V623-D637 epitopes slightly reduced the predicted score for potential epitopes. Conclusions The present study showed a very low level of genetic diversity of pvhap2 gene among P. vivax populations in the Greater Mekong Subregion. The relative conservation of pvhap2 supports further evaluation of a Pvhap2-based TBV.![]()
Collapse
Affiliation(s)
- Danni Li
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, 110122, Liaoning Province, People's Republic of China
| | - Chunyun Yu
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, 110122, Liaoning Province, People's Republic of China
| | - Jian Guo
- Department of Laboratory Medicine, Shanghai East Hospital, Tongji School of Medicine, Shanghai, People's Republic of China
| | - Yazhou Wang
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang, 110122, Liaoning Province, People's Republic of China
| | - Yan Zhao
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, 110122, Liaoning Province, People's Republic of China
| | - Lin Wang
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, 110122, Liaoning Province, People's Republic of China
| | - Myat Thu Soe
- Myanmar Health Network Organization, Yangon, Myanmar
| | - Hui Feng
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, 110122, Liaoning Province, People's Republic of China
| | | | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Lubin Jiang
- Unit of Human Parasite Molecular and Cell Biology, Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Liwang Cui
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, 3720 Spectrum Boulevard, Suite 304, Tampa, FL, 33612, USA
| | - Xiaotong Zhu
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, 110122, Liaoning Province, People's Republic of China.
| | - Yaming Cao
- Department of Immunology, College of Basic Medical Science, China Medical University, Shenyang, 110122, Liaoning Province, People's Republic of China.
| |
Collapse
|
7
|
McCaffery JN, Fonseca JA, Singh B, Cabrera-Mora M, Bohannon C, Jacob J, Arévalo-Herrera M, Moreno A. A Multi-Stage Plasmodium vivax Malaria Vaccine Candidate Able to Induce Long-Lived Antibody Responses Against Blood Stage Parasites and Robust Transmission-Blocking Activity. Front Cell Infect Microbiol 2019; 9:135. [PMID: 31119106 PMCID: PMC6504793 DOI: 10.3389/fcimb.2019.00135] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 04/15/2019] [Indexed: 12/13/2022] Open
Abstract
Malaria control and interventions including long-lasting insecticide-treated nets, indoor residual spraying, and intermittent preventative treatment in pregnancy have resulted in a significant reduction in the number of Plasmodium falciparum cases. Considerable efforts have been devoted to P. falciparum vaccines development with much less to P. vivax. Transmission-blocking vaccines, which can elicit antibodies targeting Plasmodium antigens expressed during sexual stage development and interrupt transmission, offer an alternative strategy to achieve malaria control. The post-fertilization antigen P25 mediates several functions essential to ookinete survival but is poorly immunogenic in humans. Previous clinical trials targeting this antigen have suggested that conjugation to a carrier protein could improve the immunogenicity of P25. Here we report the production, and characterization of a vaccine candidate composed of a chimeric P. vivax Merozoite Surface Protein 1 (cPvMSP1) genetically fused to P. vivax P25 (Pvs25) designed to enhance CD4+ T cell responses and its assessment in a murine model. We demonstrate that antibodies elicited by immunization with this chimeric protein recognize both the erythrocytic and sexual stages and are able to block the transmission of P. vivax field isolates in direct membrane-feeding assays. These findings provide support for the continued development of multi-stage transmission blocking vaccines targeting the life-cycle stage responsible for clinical disease and the sexual-stage development accountable for disease transmission simultaneously.
Collapse
Affiliation(s)
- Jessica N. McCaffery
- Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
| | - Jairo A. Fonseca
- Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
- Division of Infectious Diseases, Department of Medicine, Emory University, Atlanta, GA, United States
| | - Balwan Singh
- Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
| | - Monica Cabrera-Mora
- Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
| | - Caitlin Bohannon
- Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
| | - Joshy Jacob
- Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, United States
| | - Myriam Arévalo-Herrera
- Caucaseco Scientific Research Center, Malaria Vaccine and Drug Development Center, Cali, Colombia
| | - Alberto Moreno
- Emory Vaccine Center, Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
- Division of Infectious Diseases, Department of Medicine, Emory University, Atlanta, GA, United States
| |
Collapse
|
8
|
Choudhury TP, Gupta L, Kumar S. Identification, characterization and expression analysis of Anopheles stephensi double peroxidase. Acta Trop 2019; 190:210-219. [PMID: 30352205 DOI: 10.1016/j.actatropica.2018.10.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 10/13/2018] [Accepted: 10/14/2018] [Indexed: 11/28/2022]
Abstract
Peroxidases catalyze the reduction of peroxides and that, in turn, oxidize various substrates. They have been widely reported to play an important role in mosquito innate immunity against various pathogens. Here, we have characterized double heme peroxidase (AsDBLOX) gene from the Indian malaria vector Anopheles stephensi. It is a true ortholog of An. gambiae DBLOX. This 4209 bp AsDBLOX gene encodes for a protein of 1402 amino acids that has two duplicated peroxidase domains, domain I (from amino acid 61 to 527) and domain II (from amino acid 714 to 1252). The first domain has only substrate binding sites and lacks all other motifs of a functional heme peroxidase (e.g. heme binding site, calcium binding site and homodimer interface). Instead, it has two integrin binding motifs-LDV (Leu-Asp-Val) and RGD (Arg-Gly-Asp). The second peroxidase domain, however, has all the features of a complete heme peroxidase along with an integrin binding motif LDI (Leu-Asp-Ile). Thus, AsDBLOX gene is a unique type of peroxinectin as these groups of proteins are characterized by integrin binding motifs along with a heme peroxidase domain. We also observed that the AsDBLOX gene is expressed in all the life cycle stages of mosquito and is highly induced in the pupal stage of development which indicates its possible role in development.
Collapse
Affiliation(s)
- Tania Pal Choudhury
- Molecular Parasitology and Vector Biology Laboratory, Department of Biological Sciences, Birla Institute of Technology and Science (BITS), Pilani, Rajasthan, India
| | - Lalita Gupta
- Molecular Parasitology and Vector Biology Laboratory, Department of Biological Sciences, Birla Institute of Technology and Science (BITS), Pilani, Rajasthan, India; Department of Zoology, Ch. Bansi Lal University, Bhiwani, Haryana, India
| | - Sanjeev Kumar
- Molecular Parasitology and Vector Biology Laboratory, Department of Biological Sciences, Birla Institute of Technology and Science (BITS), Pilani, Rajasthan, India; Department of Biotechnology, Ch. Bansi Lal University, Bhiwani, Haryana, India.
| |
Collapse
|
9
|
Khamsehnejad MI, Djadid ND, Raz A. Identification, Molecular Characterization, and In Silico Structural Analysis of Carboxypeptidase B2 of Anopheles stephensi. JOURNAL OF MEDICAL ENTOMOLOGY 2019; 56:72-85. [PMID: 30124910 DOI: 10.1093/jme/tjy127] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Indexed: 06/08/2023]
Abstract
Malaria is a vector-borne infectious disease that is considered a priority of the World Health Organization due to its enormous impacts on global health. Plasmodium spp. (Haemosporida: Plasmodiidae), Anopheles spp. (Diptera: Culicidae), and a suitable host are the key elements for malaria transmission. To disrupt the parasitic life cycle of malaria or prevent its transmission, these three key elements should be targeted by effective control strategies. Development of vaccines that interrupt malaria transmission is one of the solutions that has been recommended to the countries that aim to eliminate malaria. With respect to the important role of Anopheles stephensi in malaria transmission and involvement of Anopheles carboxypeptidase B1 in sexual parasite development, we characterized the second member of cpb gene family (cpbAs2) of An. Stephensi to provide some basic information and evaluate significance of cpbAs2's role in complementing sexual plasmodium development role of cpbAs1. The cpbAs2 mRNA sequence was characterized by 3' and 5' RACE and the structural features of its coded protein were studied by in silico modeling. The coding sequence and gene structure of cpbAs2 were determined empirically and compared with the in silico predictions from the An. stephensi genome sequencing project. Furthermore, homology modeling revealed that its structure is very similar to the structurally important domains of procarboxypeptidase B2 in humans. This study provides basic molecular and structural information about another member of the cpb gene family of An. stephensi. The reported results are informative and necessary for evaluation of the role of this gene in sexual parasite development by future studies.
Collapse
Affiliation(s)
- Mahdokht Ilbeigi Khamsehnejad
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran (PII), Pasteur Avenue, Tehran, Iran
| | - Navid Dinparast Djadid
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran (PII), Pasteur Avenue, Tehran, Iran
| | - Abbasali Raz
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran (PII), Pasteur Avenue, Tehran, Iran
| |
Collapse
|
10
|
Martin TCS, Vinetz JM. Asymptomatic Plasmodium vivax parasitaemia in the low-transmission setting: the role for a population-based transmission-blocking vaccine for malaria elimination. Malar J 2018; 17:89. [PMID: 29466991 PMCID: PMC5822557 DOI: 10.1186/s12936-018-2243-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 02/17/2018] [Indexed: 12/21/2022] Open
Abstract
Plasmodium vivax remains an important cause of morbidity and mortality across the Americas, Horn of Africa, East and South East Asia. Control of transmission has been hampered by emergence of chloroquine resistance and several intrinsic characteristics of infection including asymptomatic carriage, challenges with diagnosis, difficulty eradicating the carrier state and early gametocyte appearance. Complex human-parasite-vector immunological interactions may facilitate onward infection of mosquitoes. Given these challenges, new therapies are being explored including the development of transmission to mosquito blocking vaccines. Herein, the case supporting the need for transmission-blocking vaccines to augment control of P. vivax parasite transmission and explore factors that are limiting eradication efforts is discussed.
Collapse
Affiliation(s)
- Thomas C S Martin
- Division of Infectious Diseases, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Joseph M Vinetz
- Division of Infectious Diseases, Department of Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
11
|
Promising approach to reducing Malaria transmission by ivermectin: Sporontocidal effect against Plasmodium vivax in the South American vectors Anopheles aquasalis and Anopheles darlingi. PLoS Negl Trop Dis 2018; 12:e0006221. [PMID: 29444080 PMCID: PMC5828505 DOI: 10.1371/journal.pntd.0006221] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 02/27/2018] [Accepted: 01/08/2018] [Indexed: 12/20/2022] Open
Abstract
Background The mosquito resistance to the insecticides threatens malaria control efforts, potentially becoming a major public health issue. Alternative methods like ivermectin (IVM) administration to humans has been suggested as a possible vector control to reduce Plasmodium transmission. Anopheles aquasalis and Anopheles darlingi are competent vectors for Plasmodium vivax, and they have been responsible for various malaria outbreaks in the coast of Brazil and the Amazon Region of South America. Methods To determine the IVM susceptibility against P. vivax in An. aquasalis and An. darlingi, ivermectin were mixed in P. vivax infected blood: (1) Powdered IVM at four concentrations (0, 5, 10, 20 or 40 ng/mL). (2) Plasma (0 hours, 4 hours, 1 day, 5, 10 and 14 days) was collected from healthy volunteers after to administer a single oral dose of IVM (200 μg/kg) (3) Mosquitoes infected with P. vivax and after 4 days was provided with IVM plasma collected 4 hours post-treatment (4) P. vivax-infected patients were treated with various combinations of IVM, chloroquine, and primaquine and plasma or whole blood was collected at 4 hours. Seven days after the infective blood meal, mosquitoes were dissected to evaluate oocyst presence. Additionally, the ex vivo effects of IVM against asexual blood-stage P. vivax was evaluated. Results IVM significantly reduced the prevalence of An. aquasalis that developed oocysts in 10 to 40 ng/mL pIVM concentrations and plasma 4 hours, 1 day and 5 days. In An. darlingi to 4 hours and 1 day. The An. aquasalis mortality was expressively increased in pIVM (40ng/mL) and plasma 4 hours, 1, 5 10 and 14 days post-intake drug and in An. darlingi only to 4 hours and 1 day. The double fed meal with mIVM by the mosquitoes has a considerable impact on the proportion of infected mosquitoes for 7 days post-feeding. The oocyst infection prevalence and intensity were notably reduced when mosquitoes ingested blood from P. vivax patients that ingested IVM+CQ, PQ+CQ and IVM+PQ+CQ. P. vivax asexual development was considerably inhibited by mIVM at four-fold dilutions. Conclusion In conclusion, whole blood spiked with IVM reduced the infection rate of P. vivax in An. aquasalis and An. darlingi, and increased the mortality of mosquitoes. Plasma from healthy volunteers after IVM administration affect asexual P. vivax development. These findings support that ivermectin may be used to decrease P. vivax transmission. Malaria is one of the most important infectious diseases in the world with hundreds of millions of new cases every year. The disease is caused by parasites of the genus Plasmodium where Plasmodium vivax represent most of the cases in the Americas. Current strategies to combat malaria transmission are being implemented; however, widespread insecticide resistance in vectors threatens the effectiveness of vector control programs. Ivermectin (IVM) has arisen as a new potential tool to be added to these programs as it has mosquito-lethal and sporontocidal properties making it a promising transmission reduction drug. Plasmodium vivax was drawn from patients, mixed with powdered IVM and metabolized IVM in plasma collected from healthy volunteers receiving IVM, and fed to mosquitoes via membrane feeding. Powdered and metabolized IVM interrupt P. vivax transmission, reducing oocyst infection and intensity rate of two South American malaria vectors An. aquasalis and An. darlingi. We also demonstrate the effect of IVM on asexual stages development of P. vivax, providing evidence that IVM may affect different parasite life cycle stages. Our findings place IVM as a strong candidate for malaria transmission reducing interventions.
Collapse
|
12
|
Menon V, Kapulu MC, Taylor I, Jewell K, Li Y, Hill F, Long CA, Miura K, Biswas S. Assessment of Antibodies Induced by Multivalent Transmission-Blocking Malaria Vaccines. Front Immunol 2018; 8:1998. [PMID: 29403479 PMCID: PMC5780346 DOI: 10.3389/fimmu.2017.01998] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 12/22/2017] [Indexed: 01/20/2023] Open
Abstract
A malaria transmission-blocking vaccine would be a critical tool in achieving malaria elimination and eradication. By using chimpanzee adenovirus serotype 63 and modified vaccinia virus Ankara viral vectored vaccines, we investigated whether incorporating two antigens into one vaccine would result in higher transmission-reducing activity than one antigen. We demonstrated that when Pfs25 was administered with other antigens Pfs28 or Pfs230C, either concurrently as a mixed vaccine or co-expressed as a dual-antigen vaccine, the antibody response in mice to each antigen was comparable to a monoantigen vaccine, without immunological interference. However, we found that the transmission-reducing activity (functional activity) of dual-antigen vaccines was not additive. Dual-antigen vaccines generally only elicited similar transmission-reducing activity to monoantigen vaccines and in one instance had lower transmission-reducing activity. We found that despite the lack of immunological interference of dual-antigen vaccines, they are still not as effective at blocking malaria transmission as Pfs25-IMX313, the current leading candidate for viral vectored vaccines. Pfs25-IMX313 elicited similar quality antibodies to dual-antigen vaccines, but higher antibody titers.
Collapse
Affiliation(s)
- Vinay Menon
- Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | - Iona Taylor
- Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Kerry Jewell
- Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Yuanyuan Li
- Jenner Institute, University of Oxford, Oxford, United Kingdom
| | | | - Carole A. Long
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Rockville, MD, United States
| | - Kazutoyo Miura
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Rockville, MD, United States
| | - Sumi Biswas
- Jenner Institute, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
13
|
Accelerated and long term stability study of Pfs25-EPA conjugates adjuvanted with Alhydrogel®. Vaccine 2017; 35:3232-3238. [PMID: 28479180 DOI: 10.1016/j.vaccine.2017.04.067] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 04/07/2017] [Accepted: 04/23/2017] [Indexed: 11/22/2022]
Abstract
Pfs25, a Plasmodium falciparum surface protein expressed during zygote and ookinete stages in infected mosquitoes, is a lead transmission-blocking vaccine candidate against falciparum malaria. To enhance immunogenicity, recombinant Pfs25 was chemically conjugated to recombinant nontoxic Pseudomonas aeruginosa ExoProtein A (rEPA) in conformance with current good manufacturing practices (cGMP), and formulated with the alum adjuvant Alhydrogel. In order to meet the regulatory requirements for a phase 1 human clinical trial, the vaccine product was extensively evaluated for stability at an initial time point and through the clinical trial period annually. Because basic quality control methods to characterize alum-based vaccines remain unavailable, a thermal forced degradation study was performed prior to the initial evaluation to identify the methods suitable to detect the quality of vaccine formulations. Our results show that the vaccine product Pfs25-EPA formulated on Alhydrogel is in conformance with regulatory guidelines and suitable for human trials.
Collapse
|
14
|
Talaat KR, Ellis RD, Hurd J, Hentrich A, Gabriel E, Hynes NA, Rausch KM, Zhu D, Muratova O, Herrera R, Anderson C, Jones D, Aebig J, Brockley S, MacDonald NJ, Wang X, Fay MP, Healy SA, Durbin AP, Narum DL, Wu Y, Duffy PE. Safety and Immunogenicity of Pfs25-EPA/Alhydrogel®, a Transmission Blocking Vaccine against Plasmodium falciparum: An Open Label Study in Malaria Naïve Adults. PLoS One 2016; 11:e0163144. [PMID: 27749907 PMCID: PMC5066979 DOI: 10.1371/journal.pone.0163144] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 09/01/2016] [Indexed: 12/31/2022] Open
Abstract
Transmission-blocking vaccines (TBVs) that target sexual stage parasite development could be an integral part of measures for malaria elimination. Pfs25 is a leading TBV candidate, and previous studies conducted in animals demonstrated an improvement of its functional immunogenicity after conjugation to EPA, a recombinant, detoxified ExoProtein A from Pseudomonas aeruginosa. In this report, we describe results of an open-label, dose-escalating Phase 1 trial to assess the safety and immunogenicity of Pfs25-EPA conjugates formulated with Alhydrogel®. Thirty malaria-naïve healthy adults received up to four doses of the conjugate vaccine, with 8, 16, or 47 μg of conjugated Pfs25 mass, at 0, 2, 4, and 10 months. Vaccinations were generally well tolerated. The majority of solicited adverse events were mild in severity with pain at the injection site the most common complaint. Anemia was the most common laboratory abnormality, but was considered possibly related to the study in only a minority of cases. No vaccine-related serious adverse events occurred. The peak geometric mean anti-Pfs25 antibody level in the highest dose group was 88 (95% CI 53, 147) μg/mL two weeks after the 4th vaccination, and declined to near baseline one year later. Antibody avidity increased over successive vaccinations. Transmission blocking activity demonstrated in a standard membrane feeding assay (SMFA) also increased from the second to the third dose, and correlated with antibody titer and, after the final dose, with antibody avidity. These results support the further evaluation of Pfs25-EPA/Alhydrogel® in a malaria-endemic population.
Collapse
Affiliation(s)
- Kawsar R. Talaat
- Center For Immunization Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Ruth D. Ellis
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Janet Hurd
- Center For Immunization Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Autumn Hentrich
- Center For Immunization Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Erin Gabriel
- Biostatistical Research Branch, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Noreen A. Hynes
- Center For Immunization Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Kelly M. Rausch
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Daming Zhu
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Olga Muratova
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Raul Herrera
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Charles Anderson
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - David Jones
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Joan Aebig
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Sarah Brockley
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Nicholas J. MacDonald
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Xiaowei Wang
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Michael P. Fay
- Biostatistical Research Branch, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Sara A. Healy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Anna P. Durbin
- Center For Immunization Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - David L. Narum
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Yimin Wu
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
- * E-mail:
| | - Patrick E. Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| |
Collapse
|
15
|
Reiner RC, Guerra C, Donnelly MJ, Bousema T, Drakeley C, Smith DL. Estimating malaria transmission from humans to mosquitoes in a noisy landscape. J R Soc Interface 2016; 12:20150478. [PMID: 26400195 PMCID: PMC4614487 DOI: 10.1098/rsif.2015.0478] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
A basic quantitative understanding of malaria transmission requires measuring the probability a mosquito becomes infected after feeding on a human. Parasite prevalence in mosquitoes is highly age-dependent, and the unknown age-structure of fluctuating mosquito populations impedes estimation. Here, we simulate mosquito infection dynamics, where mosquito recruitment is modelled seasonally with fractional Brownian noise, and we develop methods for estimating mosquito infection rates. We find that noise introduces bias, but the magnitude of the bias depends on the ‘colour' of the noise. Some of these problems can be overcome by increasing the sampling frequency, but estimates of transmission rates (and estimated reductions in transmission) are most accurate and precise if they combine parity, oocyst rates and sporozoite rates. These studies provide a basis for evaluating the adequacy of various entomological sampling procedures for measuring malaria parasite transmission from humans to mosquitoes and for evaluating the direct transmission-blocking effects of a vaccine.
Collapse
Affiliation(s)
- Robert C Reiner
- Fogarty International Center, National Institutes of Health, Bethesda, MD, USA Department of Entomology, University of California, Davis, CA, USA Department of Epidemiology and Biostatistics, Indiana University, Bloomington, IN, USA
| | - Carlos Guerra
- Center for Disease Dynamics, Economics and Policy, Washington, DC, USA
| | - Martin J Donnelly
- Department of Vector Biology, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, UK Malaria Programme, Wellcome Trust Sanger Institute, Cambridge, UK
| | - Teun Bousema
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, UK Department of Medical Microbiology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Chris Drakeley
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, UK
| | - David L Smith
- Fogarty International Center, National Institutes of Health, Bethesda, MD, USA Sanaria Institute for Global Health and Tropical Medicine, Rockville, MD, USA Department of Zoology, University of Oxford, Oxford, UK Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, USA
| |
Collapse
|
16
|
Dunachie S, Hill AVS, Fletcher HA. Profiling the host response to malaria vaccination and malaria challenge. Vaccine 2015; 33:5316-20. [PMID: 26256528 PMCID: PMC4582768 DOI: 10.1016/j.vaccine.2015.07.107] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 07/27/2015] [Accepted: 07/29/2015] [Indexed: 12/21/2022]
Abstract
A vaccine for malaria is urgently required. The RTS,S vaccine represents major progress, but is only partially effective. Development of the next generation of highly effective vaccines requires elucidation of the protective immune response. Immunity to malaria is known to be complex, and pattern-based approaches such as global gene expression profiling are ideal for understanding response to vaccination and protection against disease. The availability of experimental sporozoite challenge in humans to test candidate malaria vaccines offers a precious opportunity unavailable for other current targets of vaccine research such as HIV, tuberculosis and Ebola. However, a limited number of transcriptional profiling studies in the context of malaria vaccine research have been published to date. This review outlines the background, existing studies, limits and opportunities for gene expression studies to accelerate malaria vaccine research.
Collapse
Affiliation(s)
- Susanna Dunachie
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Churchill Hospital, Oxford OX3 7LJ, UK; Mahidol-Oxford Tropical Medicine Research Unit, 3rd Floor, 60th Anniversary Chalermprakiat Building, 420/6 Ratchawithi Road, Bangkok 10400, Thailand; Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine Research Building, University of Oxford, Old Road Campus, Roosevelt Drive, Oxford OX3 7FZ, UK.
| | - Adrian V S Hill
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Churchill Hospital, Oxford OX3 7LJ, UK
| | - Helen A Fletcher
- London School of Hygiene & Tropical Medicine, London, W1CE 7HT, UK; The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Churchill Hospital, Oxford OX3 7LJ, UK
| |
Collapse
|
17
|
Sauerwein RW, Bousema T. Transmission blocking malaria vaccines: Assays and candidates in clinical development. Vaccine 2015; 33:7476-82. [PMID: 26409813 DOI: 10.1016/j.vaccine.2015.08.073] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 07/31/2015] [Accepted: 08/05/2015] [Indexed: 12/20/2022]
Abstract
Stimulated by recent advances in malaria control and increased funding, the elimination of malaria is now considered to be an attainable goal for an increasing number of malaria-endemic regions. This has boosted the interest in transmission-reducing interventions including vaccines that target sexual, sporogenic, and/or mosquito-stage antigens to interrupt malaria transmission (SSM-VIMT). SSM-VIMT aim to prevent human malaria infection in vaccinated communities by inhibiting parasite development within the mosquito after a blood meal taken from a gametocyte carrier. Only a handful of target antigens are in clinical development and progress has been slow over the years. Major stumbling blocks include (i) the expression of appropriately folded target proteins and their downstream purification, (ii) insufficient induction of sustained functional blocking antibody titers by candidate vaccines in humans, and (iii) validation of a number of (bio)-assays as correlate for blocking activity in the field. Here we discuss clinical manufacturing and testing of current SSM-VIMT candidates and the latest bio-assay development for clinical evaluation. New testing strategies are discussed that may accelerate the evaluation and application of SSM-VIMT.
Collapse
Affiliation(s)
- R W Sauerwein
- Department of Medical Microbiology, Radboud University Medical Center, PO Box 9101 (268), Geert Grooteplein 28, 6500 HB Nijmegen, The Netherlands.
| | - T Bousema
- Department of Medical Microbiology, Radboud University Medical Center, PO Box 9101 (268), Geert Grooteplein 28, 6500 HB Nijmegen, The Netherlands
| |
Collapse
|
18
|
Beiss V, Spiegel H, Boes A, Kapelski S, Scheuermayer M, Edgue G, Sack M, Fendel R, Reimann A, Schillberg S, Pradel G, Fischer R. Heat-precipitation allows the efficient purification of a functional plant-derived malaria transmission-blocking vaccine candidate fusion protein. Biotechnol Bioeng 2015; 112:1297-305. [PMID: 25615702 DOI: 10.1002/bit.25548] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Revised: 01/06/2015] [Accepted: 01/15/2015] [Indexed: 12/22/2022]
Abstract
Malaria is a vector-borne disease affecting more than two million people and accounting for more than 600,000 deaths each year, especially in developing countries. The most serious form of malaria is caused by Plasmodium falciparum. The complex life cycle of this parasite, involving pre-erythrocytic, asexual and sexual stages, makes vaccine development cumbersome but also offers a broad spectrum of vaccine candidates targeting exactly those stages. Vaccines targeting the sexual stage of P. falciparum are called transmission-blocking vaccines (TBVs). They do not confer protection for the vaccinated individual but aim to reduce or prevent the transmission of the parasite within a population and are therefore regarded as an essential tool in the fight against the disease. Malaria predominantly affects large populations in developing countries, so TBVs need to be produced in large quantities at low cost. Combining the advantages of eukaryotic expression with a virtually unlimited upscaling potential and a good product safety profile, plant-based expression systems represent a suitable alternative for the production of TBVs. We report here the high level (300 μg/g fresh leaf weight (FLW)) transient expression in Nicotiana benthamiana leaves of an effective TBV candidate based on a fusion protein F0 comprising Pfs25 and the C0-domain of Pfs230, and the implementation of a simple and cost-effective heat treatment step for purification that yields intact recombinant protein at >90% purity with a recovery rate of >70%. The immunization of mice clearly showed that antibodies raised against plant-derived F0 completely blocked the formation of oocysts in a malaria transmission-blocking assay (TBA) making F0 an interesting TBV candidate or a component of a multi-stage malaria vaccine cocktail.
Collapse
Affiliation(s)
- Veronique Beiss
- Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Forckenbeckstrasse 6, 52074, Aachen, Germany
| | - Holger Spiegel
- Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Forckenbeckstrasse 6, 52074, Aachen, Germany
| | - Alexander Boes
- Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Forckenbeckstrasse 6, 52074, Aachen, Germany.
| | - Stephanie Kapelski
- Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Forckenbeckstrasse 6, 52074, Aachen, Germany
| | - Matthias Scheuermayer
- Research Center for Infectious Diseases, University of Wuerzburg, Josef Schneider Str. 2/Bau D15, 97080, Wuerzburg, Germany
| | - Gueven Edgue
- Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Forckenbeckstrasse 6, 52074, Aachen, Germany
| | - Markus Sack
- Institute for Molecular Biotechnology, RWTH Aachen University, Worringer Weg 1, 52074, Aachen, Germany
| | - Rolf Fendel
- Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Forckenbeckstrasse 6, 52074, Aachen, Germany
| | - Andreas Reimann
- Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Forckenbeckstrasse 6, 52074, Aachen, Germany
| | - Stefan Schillberg
- Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Forckenbeckstrasse 6, 52074, Aachen, Germany
| | - Gabriele Pradel
- Research Center for Infectious Diseases, University of Wuerzburg, Josef Schneider Str. 2/Bau D15, 97080, Wuerzburg, Germany
- Institute for Molecular Biotechnology, RWTH Aachen University, Worringer Weg 1, 52074, Aachen, Germany
| | - Rainer Fischer
- Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Forckenbeckstrasse 6, 52074, Aachen, Germany
- Institute for Molecular Biotechnology, RWTH Aachen University, Worringer Weg 1, 52074, Aachen, Germany
| |
Collapse
|
19
|
Nikolaeva D, Draper SJ, Biswas S. Toward the development of effective transmission-blocking vaccines for malaria. Expert Rev Vaccines 2015; 14:653-80. [PMID: 25597923 DOI: 10.1586/14760584.2015.993383] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The continued global burden of malaria can in part be attributed to a complex lifecycle, with both human hosts and mosquito vectors serving as transmission reservoirs. In preclinical models of vaccine-induced immunity, antibodies to parasite sexual-stage antigens, ingested in the mosquito blood meal, can inhibit parasite survival in the insect midgut as judged by ex vivo functional studies such as the membrane feeding assay. In an era of renewed political momentum for malaria elimination and eradication campaigns, such observations have fueled support for the development and implementation of so-called transmission-blocking vaccines. While leading candidates are being evaluated using a variety of promising vaccine platforms, the field is also beginning to capitalize on global '-omics' data for the rational genome-based selection and unbiased characterization of parasite and mosquito proteins to expand the candidate list. This review covers the progress and prospects of these recent developments.
Collapse
Affiliation(s)
- Daria Nikolaeva
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Headington, Oxford OX3 7DQ, UK
| | | | | |
Collapse
|
20
|
Da DF, Churcher TS, Yerbanga RS, Yaméogo B, Sangaré I, Ouedraogo JB, Sinden RE, Blagborough AM, Cohuet A. Experimental study of the relationship between Plasmodium gametocyte density and infection success in mosquitoes; implications for the evaluation of malaria transmission-reducing interventions. Exp Parasitol 2014; 149:74-83. [PMID: 25541384 DOI: 10.1016/j.exppara.2014.12.010] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 12/10/2014] [Accepted: 12/15/2014] [Indexed: 11/29/2022]
Abstract
The evaluation of transmission reducing interventions (TRI) to control malaria widely uses membrane feeding assays. In such assays, the intensity of Plasmodium infection in the vector might affect the measured efficacy of the candidates to block transmission. Gametocyte density in the host blood is a determinant of the infection success in the mosquito, however, uncertain estimates of parasite densities and intrinsic characteristics of the infected blood can induce variability. To reduce this variation, a feasible method is to dilute infectious blood samples. We describe the effect of diluting samples of Plasmodium-containing blood samples to allow accurate relative measures of gametocyte densities and their impact on mosquito infectivity and TRI efficacy. Natural Plasmodium falciparum samples were diluted to generate a wide range of parasite densities, and fed to Anopheles coluzzii mosquitoes. This was compared with parallel dilutions conducted on Plasmodium berghei infections. We examined how blood dilution influences the observed blocking activity of anti-Pbs28 monoclonal antibody using the P. berghei/Anopheles stephensi system. In the natural species combination P. falciparum/An. coluzzii, blood dilution using heat-inactivated, infected blood as diluents, revealed positive near linear relationships, between gametocyte densities and oocyst loads in the range tested. A similar relationship was observed in the P. berghei/An. stephensi system when using a similar dilution method. In contrast, diluting infected mice blood with fresh uninfected blood dramatically increases the infectiousness. This suggests that highly infected mice blood contains inhibitory factors or reduced blood moieties, which impede infection and may in turn, lead to misinterpretation when comparing individual TRI evaluation assays. In the lab system, the transmission blocking activity of an antibody specific for Pbs28 was confirmed to be density-dependent. This highlights the need to carefully interpret evaluations of TRI candidates, regarding gametocyte densities in the P. berghei/An. stephensi system.
Collapse
Affiliation(s)
- Dari F Da
- Institut de Recherche en Sciences de la Santé, Direction Régionale, 399 avenue de la liberté, Bobo Dioulasso 01 01 BP 545, Burkina Faso; Institut de Recherche pour le Développement, unité MIVEGEC (UM1-UM2-CNRS 5290-IRD 224), 911 avenue Agropolis, Montpellier Cedex 5 34394, France
| | - Thomas S Churcher
- Department of Infectious Disease Epidemiology, Imperial College London, London, United Kingdom
| | - Rakiswendé S Yerbanga
- Institut de Recherche en Sciences de la Santé, Direction Régionale, 399 avenue de la liberté, Bobo Dioulasso 01 01 BP 545, Burkina Faso
| | - Bienvenue Yaméogo
- Institut de Recherche en Sciences de la Santé, Direction Régionale, 399 avenue de la liberté, Bobo Dioulasso 01 01 BP 545, Burkina Faso
| | - Ibrahim Sangaré
- Institut de Recherche en Sciences de la Santé, Direction Régionale, 399 avenue de la liberté, Bobo Dioulasso 01 01 BP 545, Burkina Faso; Institut de Recherche pour le Développement, unité MIVEGEC (UM1-UM2-CNRS 5290-IRD 224), 911 avenue Agropolis, Montpellier Cedex 5 34394, France
| | - Jean Bosco Ouedraogo
- Institut de Recherche en Sciences de la Santé, Direction Régionale, 399 avenue de la liberté, Bobo Dioulasso 01 01 BP 545, Burkina Faso
| | - Robert E Sinden
- Division of Cell and Molecular Biology, Imperial College London, London, United Kingdom; The Jenner Institute, University of Oxford, Roosevelt Drive, Oxford OX3 7DQ, United Kingdom
| | - Andrew M Blagborough
- Division of Cell and Molecular Biology, Imperial College London, London, United Kingdom
| | - Anna Cohuet
- Institut de Recherche en Sciences de la Santé, Direction Régionale, 399 avenue de la liberté, Bobo Dioulasso 01 01 BP 545, Burkina Faso; Institut de Recherche pour le Développement, unité MIVEGEC (UM1-UM2-CNRS 5290-IRD 224), 911 avenue Agropolis, Montpellier Cedex 5 34394, France.
| |
Collapse
|
21
|
da Costa M, Pinheiro-Silva R, Antunes S, Moreno-Cid JA, Custódio A, Villar M, Silveira H, de la Fuente J, Domingos A. Mosquito Akirin as a potential antigen for malaria control. Malar J 2014; 13:470. [PMID: 25472895 PMCID: PMC4265507 DOI: 10.1186/1475-2875-13-470] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 11/27/2014] [Indexed: 11/10/2022] Open
Abstract
Background The control of vector-borne diseases is important to improve human and animal health worldwide. Malaria is one of the world’s deadliest diseases and is caused by protozoan parasites of the genus Plasmodium, which are transmitted by Anopheles spp. mosquitoes. Recent evidences using Subolesin (SUB) and Akirin (AKR) vaccines showed a reduction in the survival and/or fertility of blood-sucking ectoparasite vectors and the infection with vector-borne pathogens. These experiments suggested the possibility of using AKR for malaria control. Methods The role of AKR on Plasmodium berghei infection and on the fitness and reproduction of the main malaria vector, Anopheles gambiae was characterized by evaluating the effect of akr gene knockdown or vaccination with recombinant mosquito AKR on parasite infection levels, fertility and mortality of female mosquitoes. Results Gene knockdown by RNA interference in mosquitoes suggested a role for akr in mosquito survival and fertility. Vaccination with recombinant Aedes albopictus AKR reduced parasite infection in mosquitoes fed on immunized mice when compared to controls. Conclusions These results showed that recombinant AKR could be used to develop vaccines for malaria control. If effective, AKR-based vaccines could be used to immunize wildlife reservoir hosts and/or humans to reduce the risk of pathogen transmission. However, these vaccines need to be evaluated under field conditions to characterize their effect on vector populations and pathogen infection and transmission.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Ana Domingos
- Instituto de Higiene e Medicina Tropical, Rua da Junqueira 100, 1349-008 Lisbon, Portugal.
| |
Collapse
|
22
|
Zhao R, Mohammed-Awel J. A mathematical model studying mosquito-stage transmission-blocking vaccines. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2014; 11:1229-1245. [PMID: 25347811 DOI: 10.3934/mbe.2014.11.1229] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
A compartmental deterministic model is proposed to evaluate the effectiveness of transmission-blocking vaccines of malaria, which targets at the parasite stage in the mosquito. The model is rigorously analyzed and numerical simulations are performed. The results and implications are discussed.
Collapse
Affiliation(s)
- Ruijun Zhao
- Department of Mathematics and Statistics, Minnesota State University, Mankato, Mankaot, MN, 56001, United States.
| | | |
Collapse
|
23
|
Mathematical analysis of an age-structured model for malaria transmission dynamics. Math Biosci 2014; 247:80-94. [DOI: 10.1016/j.mbs.2013.10.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Revised: 09/16/2013] [Accepted: 10/31/2013] [Indexed: 11/18/2022]
|
24
|
Shimp RL, Rowe C, Reiter K, Chen B, Nguyen V, Aebig J, Rausch KM, Kumar K, Wu Y, Jin AJ, Jones DS, Narum DL. Development of a Pfs25-EPA malaria transmission blocking vaccine as a chemically conjugated nanoparticle. Vaccine 2013; 31:2954-62. [PMID: 23623858 PMCID: PMC3683851 DOI: 10.1016/j.vaccine.2013.04.034] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Revised: 02/28/2013] [Accepted: 04/11/2013] [Indexed: 11/16/2022]
Abstract
Successful efforts to control infectious diseases have often required the use of effective vaccines. The current global strategy for control of malaria, including elimination and eradication will also benefit from the development of an effective vaccine that interrupts malaria transmission. To this end, a vaccine that disrupts malaria transmission within the mosquito host has been investigated for several decades targeting a 25 kDa ookinete specific surface protein, identified as Pfs25. Phase 1 human trial results using a recombinant Pfs25H/Montanide ISA51 formulation demonstrated that human Pfs25 specific antibodies block parasite infectivity to mosquitoes; however, the extent of blocking was likely insufficient for an effective transmission blocking vaccine. To overcome the poor immunogenicity, processes to produce and characterize recombinant Pfs25H conjugated to a detoxified form of Pseudomonas aeruginosa exoprotein A (EPA) have been developed and used to manufacture a cGMP pilot lot for use in human clinical trials. The Pfs25-EPA conjugate appears as a nanoparticle with an average molar mass in solution of approximately 600 kDa by static light scattering with an average diameter 20 nm (range 10-40 nm) by dynamic light scattering. The molar ratio of Pfs25H to EPA is about 3 to 1 by amino acid analysis, respectively. Outbred mice immunized with the Pfs25-EPA conjugated nanoparticle formulated on Alhydrogel(®) had a 75-110 fold increase in Pfs25H specific antibodies when compared to an unconjugated Pfs25H/Alhydrogel(®) formulation. A phase 1 human trial using the Pfs25-EPA/Alhydrogel(®) formulation is ongoing in the United States.
Collapse
Affiliation(s)
- Richard L. Shimp
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Disease, National Institutes of Health, Rockville, MD 20852, USA
| | - Christopher Rowe
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Disease, National Institutes of Health, Rockville, MD 20852, USA
| | - Karine Reiter
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Disease, National Institutes of Health, Rockville, MD 20852, USA
| | - Beth Chen
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Disease, National Institutes of Health, Rockville, MD 20852, USA
| | - Vu Nguyen
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Disease, National Institutes of Health, Rockville, MD 20852, USA
| | - Joan Aebig
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Disease, National Institutes of Health, Rockville, MD 20852, USA
| | - Kelly M. Rausch
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Disease, National Institutes of Health, Rockville, MD 20852, USA
| | - Krishan Kumar
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Disease, National Institutes of Health, Rockville, MD 20852, USA
| | - Yimin Wu
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Disease, National Institutes of Health, Rockville, MD 20852, USA
| | - Albert J. Jin
- Laboratory of Cellular Imaging and Macromolecular Biophysics, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, USA
| | - David S. Jones
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Disease, National Institutes of Health, Rockville, MD 20852, USA
| | - David L. Narum
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Disease, National Institutes of Health, Rockville, MD 20852, USA
| |
Collapse
|
25
|
Riley EM, Stewart VA. Immune mechanisms in malaria: new insights in vaccine development. Nat Med 2013; 19:168-78. [DOI: 10.1038/nm.3083] [Citation(s) in RCA: 144] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Accepted: 01/07/2013] [Indexed: 02/07/2023]
|
26
|
Crawford JE, Rottschaefer SM, Coulibaly B, Sacko M, Niaré O, Riehle MM, Traore SF, Vernick KD, Lazzaro BP. No evidence for positive selection at two potential targets for malaria transmission-blocking vaccines in Anopheles gambiae s.s. INFECTION GENETICS AND EVOLUTION 2013; 16:87-92. [PMID: 23357581 DOI: 10.1016/j.meegid.2013.01.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Revised: 01/04/2013] [Accepted: 01/05/2013] [Indexed: 12/27/2022]
Abstract
Human malaria causes nearly a million deaths in sub-Saharan Africa each year. The evolution of drug-resistance in the parasite and insecticide resistance in the mosquito vector has complicated control measures and made the need for new control strategies more urgent. Anopheles gambiae s.s. is one of the primary vectors of human malaria in Africa, and parasite-transmission-blocking vaccines targeting Anopheles proteins have been proposed as a possible strategy to control the spread of the disease. However, the success of these hypothetical technologies would depend on the successful ability to broadly target mosquito populations that may be genetically heterogeneous. Understanding the evolutionary pressures shaping genetic variation among candidate target molecules offers a first step towards evaluating the prospects of successfully deploying such technologies. We studied the population genetics of genes encoding two candidate target proteins, the salivary gland protein saglin and the basal lamina structural protein laminin, in wild populations of the M and S molecular forms of A. gambiae in Mali. Through analysis of intraspecific genetic variation and interspecific comparisons, we found no evidence of positive natural selection at the genes encoding these proteins. On the contrary, we found evidence for particularly strong purifying selection at the laminin gene. These results provide insight into the patterns of genetic diversity of saglin and laminin, and we discuss these findings in relation to the potential development of these molecules as vaccine targets.
Collapse
|
27
|
Control of multiple arthropod vector infestations with subolesin/akirin vaccines. Vaccine 2013; 31:1187-96. [PMID: 23291476 DOI: 10.1016/j.vaccine.2012.12.073] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Revised: 12/20/2012] [Accepted: 12/23/2012] [Indexed: 11/24/2022]
Abstract
Diseases transmitted by arthropod vectors such as mosquitoes, ticks and sand flies greatly impact human and animal health and thus their control is important for the eradication of vector-borne diseases (VBD). Vaccination is an environmentally friendly alternative for vector control that allows control of several VBD by targeting their common vector. Recent results have suggested that subolesin/akirin (SUB/AKR) is good candidate antigens for the control of arthropod vector infestations. Here we describe the comparative effect of vaccination with SUB, AKR and Q38 and Q41 chimeras containing SUB/AKR conserved protective epitopes on tick, mosquitoes and sand flies vector mortality, molting, oviposition and/or fertility. We demonstrated that SUB vaccination had the highest efficacy (E) across all vector species (54-92%), Q41 vaccination had the highest vaccine E in mosquitoes (99%) by reducing female survival and fertility, and Q38 vaccination had the highest effect on reducing mosquito (28%) and sand fly (26%) oviposition. The effect of vaccination on different developmental processes in several important arthropod vectors encourages the development of SUB/AKR universal vaccines for the control of multiple vector infestations and reduction of VBD.
Collapse
|
28
|
McCoy JM, Tonkin CJ. Calcium and repression in malaria sex: knowing when the time is right. Cell Host Microbe 2013; 12:1-2. [PMID: 22817981 DOI: 10.1016/j.chom.2012.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Translational repression is important for development of the malaria parasite when establishing infection in the mosquito. In this issue of Cell Host and Microbe, Sebastian et al. (2012) show that a calcium-dependent protein kinase is important for alleviating translational repression during developmental progression.
Collapse
Affiliation(s)
- James M McCoy
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | | |
Collapse
|
29
|
Jones CS, Luong T, Hannon M, Tran M, Gregory JA, Shen Z, Briggs SP, Mayfield SP. Heterologous expression of the C-terminal antigenic domain of the malaria vaccine candidate Pfs48/45 in the green algae Chlamydomonas reinhardtii. Appl Microbiol Biotechnol 2012; 97:1987-95. [DOI: 10.1007/s00253-012-4071-7] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Revised: 03/28/2012] [Accepted: 03/29/2012] [Indexed: 12/18/2022]
|
30
|
Angrisano F, Tan YH, Sturm A, McFadden GI, Baum J. Malaria parasite colonisation of the mosquito midgut – Placing the Plasmodium ookinete centre stage. Int J Parasitol 2012; 42:519-27. [DOI: 10.1016/j.ijpara.2012.02.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Revised: 02/03/2012] [Accepted: 02/04/2012] [Indexed: 11/28/2022]
|
31
|
Bounkeua V, Li F, Chuquiyauri R, Abeles SR, McClean CM, Neyra V, Llanos-Cuentas A, Yori PP, Vinetz JM. Lack of molecular correlates of Plasmodium vivax ookinete development. Am J Trop Med Hyg 2011; 85:207-13. [PMID: 21813836 DOI: 10.4269/ajtmh.2011.10-0729] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Previous studies of Plasmodium vivax transmission to Anopheles spp. mosquitoes have not been able to predict mosquito infectivity on the basis of microscopic or molecular quantification of parasites (total parasites in the sample or total number of gametocytes) in infected blood. Two methods for production of P. vivax ookinete cultures in vitro, with yields of 10(6) macrogametocytes, 10(4) zygotes, and 10(3) ookinetes, respectively, per 10 mL of P. vivax-infected patient blood with approximately 0.01% parasitemia, were used to study P. vivax sexual stage development. The quantity of gametocytes, determined by counting Giemsa-stained blood smears, and quantity and type of gametocyte as determined by quantitative reverse transcriptase-polymerase chain reaction for Pvalpha tubulin II and macrogametocyte-specific pvg377 did not predict ookinete yield. Factors that affect the efficiency of in vitro P. vivax ookinete transformation remain poorly understood.
Collapse
Affiliation(s)
- Viengngeun Bounkeua
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
de la Fuente J, Moreno-Cid JA, Canales M, Villar M, de la Lastra JMP, Kocan KM, Galindo RC, Almazán C, Blouin EF. Targeting arthropod subolesin/akirin for the development of a universal vaccine for control of vector infestations and pathogen transmission. Vet Parasitol 2011; 181:17-22. [DOI: 10.1016/j.vetpar.2011.04.018] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
33
|
Noden BH, Vaughan JA, Pumpuni CB, Beier JC. Mosquito ingestion of antibodies against mosquito midgut microbiota improves conversion of ookinetes to oocysts for Plasmodium falciparum, but not P. yoelii. Parasitol Int 2011; 60:440-6. [PMID: 21763778 DOI: 10.1016/j.parint.2011.07.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2011] [Revised: 06/07/2011] [Accepted: 07/04/2011] [Indexed: 11/25/2022]
Abstract
The mosquito midgut is a site of complex interactions between the mosquito, the malaria parasite and the resident bacterial flora. In laboratory experiments, we observed significant enhancement of Plasmodium falciparum oocyst production when Anopheles gambiae (Diptera: Culicidae) mosquitoes were membrane-fed on infected blood containing gametocytes from in vitro cultures mixed with sera from rabbits immunized with A. gambiae midguts. To identify specific mechanisms, we evaluated whether the immune sera was interfering with the usual limiting activity of gram-negative bacteria in An. gambiae midguts. Enhancement of P. falciparum infection rates occurred at some stage between the ookinete and oocyst stage and was associated with greater numbers of oocysts in mosquitoes fed on immune sera. The same immune sera did not affect the sporogonic development of P. yoelii, a rodent malaria parasite. Not only did antibodies in the immune sera recognize several types of midgut-derived gram-negative bacteria (Pseudomonas spp. and Cedecea spp.), but gentamicin provided in the sugar meal 3 days before an infectious P. falciparum blood meal mixed with immune sera eliminated the enhancing effect. These results suggest that gram-negative bacteria, which normally impair P. falciparum development between the ookinete and oocyst stage, were altered by specific anti-bacterial antibodies produced by immunizing rabbits with non-antibiotic-treated midgut lysates. Because of the differences in developmental kinetics between human and rodent malaria species, the anti-bacterial antibodies had no effect on P. yoelii because their ookinetes leave the midgut much earlier than P. falciparum and so are not influenced as strongly by resident midgut bacteria. While this study highlights the complex interactions occurring between the parasite, mosquito, and midgut microbiota, the ultimate goal is to determine the influence of midgut microbiota on Plasmodium development in anopheline midguts in malaria endemic settings.
Collapse
Affiliation(s)
- Bruce H Noden
- Department of Biomedical Science, School of Health and Applied Sciences, Polytechnic of Namibia, Windhoek, Namibia.
| | | | | | | |
Collapse
|
34
|
Smith TA, Chitnis N, Briët OJ, Tanner M. Uses of mosquito-stage transmission-blocking vaccines against Plasmodium falciparum. Trends Parasitol 2011; 27:190-6. [DOI: 10.1016/j.pt.2010.12.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Revised: 12/27/2010] [Accepted: 12/30/2010] [Indexed: 11/29/2022]
|
35
|
Bousema T, Drakeley C. Epidemiology and infectivity of Plasmodium falciparum and Plasmodium vivax gametocytes in relation to malaria control and elimination. Clin Microbiol Rev 2011; 24:377-410. [PMID: 21482730 PMCID: PMC3122489 DOI: 10.1128/cmr.00051-10] [Citation(s) in RCA: 508] [Impact Index Per Article: 39.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Malaria remains a major cause of morbidity and mortality in the tropics, with Plasmodium falciparum responsible for the majority of the disease burden and P. vivax being the geographically most widely distributed cause of malaria. Gametocytes are the sexual-stage parasites that infect Anopheles mosquitoes and mediate the onward transmission of the disease. Gametocytes are poorly studied despite this crucial role, but with a recent resurgence of interest in malaria elimination, the study of gametocytes is in vogue. This review highlights the current state of knowledge with regard to the development and longevity of P. falciparum and P. vivax gametocytes in the human host and the factors influencing their distribution within endemic populations. The evidence for immune responses, antimalarial drugs, and drug resistance influencing infectiousness to mosquitoes is reviewed. We discuss how the application of molecular techniques has led to the identification of submicroscopic gametocyte carriage and to a reassessment of the human infectious reservoir. These components are drawn together to show how control measures that aim to reduce malaria transmission, such as mass drug administration and a transmission-blocking vaccine, might better be deployed.
Collapse
Affiliation(s)
- Teun Bousema
- Department of Immunology & Infection, London School of Hygiene and Tropical Medicine, London W1CE 7HT, United Kingdom
| | - Chris Drakeley
- Department of Immunology & Infection, London School of Hygiene and Tropical Medicine, London W1CE 7HT, United Kingdom
| |
Collapse
|
36
|
Torres L, Almazán C, Ayllón N, Galindo RC, Rosario-Cruz R, Quiroz-Romero H, de la Fuente J. Functional genomics of the horn fly, Haematobia irritans (Linnaeus, 1758). BMC Genomics 2011; 12:105. [PMID: 21310032 PMCID: PMC3045961 DOI: 10.1186/1471-2164-12-105] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2010] [Accepted: 02/10/2011] [Indexed: 12/27/2022] Open
Abstract
Background The horn fly, Haematobia irritans (Linnaeus, 1758) (Diptera: Muscidae) is one of the most important ectoparasites of pastured cattle. Horn flies infestations reduce cattle weight gain and milk production. Additionally, horn flies are mechanical vectors of different pathogens that cause disease in cattle. The aim of this study was to conduct a functional genomics study in female horn flies using Expressed Sequence Tags (EST) analysis and RNA interference (RNAi). Results A cDNA library was made from whole abdominal tissues collected from partially fed adult female horn flies. High quality horn fly ESTs (2,160) were sequenced and assembled into 992 unigenes (178 contigs and 814 singlets) representing molecular functions such as serine proteases, cell metabolism, mitochondrial function, transcription and translation, transport, chromatin structure, vitellogenesis, cytoskeleton, DNA replication, cell response to stress and infection, cell proliferation and cell-cell interactions, intracellular trafficking and secretion, and development. Functional analyses were conducted using RNAi for the first time in horn flies. Gene knockdown by RNAi resulted in higher horn fly mortality (protease inhibitor functional group), reduced oviposition (vitellogenin, ferritin and vATPase groups) or both (immune response and 5'-NUC groups) when compared to controls. Silencing of ubiquitination ESTs did not affect horn fly mortality and ovisposition while gene knockdown in the ferritin and vATPse functional groups reduced mortality when compared to controls. Conclusions These results advanced the molecular characterization of this important ectoparasite and suggested candidate protective antigens for the development of vaccines for the control of horn fly infestations.
Collapse
Affiliation(s)
- Lorena Torres
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma de Tamaulipas, Km, 5 carretera Victoria-Mante, CP 87000 Ciudad Victoria, Tamaulipas, Mexico
| | | | | | | | | | | | | |
Collapse
|
37
|
Dessens JT, Saeed S, Tremp AZ, Carter V. Malaria crystalloids: specialized structures for parasite transmission? Trends Parasitol 2011; 27:106-10. [PMID: 21237711 PMCID: PMC3133641 DOI: 10.1016/j.pt.2010.12.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2010] [Revised: 12/13/2010] [Accepted: 12/14/2010] [Indexed: 11/22/2022]
Abstract
Malaria parasites possess many unique subcellular structures and organelles that are essential for the successful completion of the complex life cycle of Plasmodium in the vertebrate host and mosquito vector. Among these are the crystalloids: transient structures whose presence is restricted to the mosquito-specific ookinete and young oocyst stages of the parasite. Nearly five decades after they were first described, the crystalloids are back in the spotlight, with recent discoveries pointing to an important role in protein trafficking and sporozoite transmission that could be exploited as new targets for control of malaria transmission.
Collapse
Affiliation(s)
- Johannes T Dessens
- Department of Pathogen Molecular Biology, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom.
| | | | | | | |
Collapse
|
38
|
Bounkeua V, Li F, Vinetz JM. In vitro generation of Plasmodium falciparum ookinetes. Am J Trop Med Hyg 2011; 83:1187-94. [PMID: 21118920 DOI: 10.4269/ajtmh.2010.10-0433] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Plasmodium transmission from the human host to the mosquito depends on the ability of gametocytes to differentiate into ookinetes, the invasive form of the parasite that invades and establishes infection in the mosquito midgut. The biology of P. falciparum ookinetes is poorly understood, because sufficient quantities of this stage of this parasite species have not been obtained for detailed study. This report details methods to optimize production of P. falciparum sexual stage parasites, including ookinetes. Flow cytometric sorting was used to separate diploid/tetraploid zygotes and ookinetes from haploid gametetocytes and unfertilized gametes based on DNA content. Consistent production of 10(6)-10(7) P. falciparum ookinetes per 10 mL culture was observed, with ookinete transformation present in 10-40% of all parasite forms. Transmission electron micrographs of cultured parasites confirmed ookinete development.
Collapse
Affiliation(s)
- Viengngeun Bounkeua
- Division of Infectious Diseases, Department of Medicine, University of California San Diego, La Jolla, California 92093-0741, USA
| | | | | |
Collapse
|
39
|
Moreno-Cid JA, Jiménez M, Cornelie S, Molina R, Alarcón P, Lacroix MN, Pinal R, Delacour S, Lucientes J, Canales M, de la Lastra JMP, Villar M, de la Fuente J. Characterization of Aedes albopictus akirin for the control of mosquito and sand fly infestations. Vaccine 2010; 29:77-82. [DOI: 10.1016/j.vaccine.2010.10.011] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2010] [Revised: 09/24/2010] [Accepted: 10/06/2010] [Indexed: 11/25/2022]
|
40
|
Churcher TS, Dawes EJ, Sinden RE, Christophides GK, Koella JC, Basáñez MG. Population biology of malaria within the mosquito: density-dependent processes and potential implications for transmission-blocking interventions. Malar J 2010; 9:311. [PMID: 21050427 PMCID: PMC2988043 DOI: 10.1186/1475-2875-9-311] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2010] [Accepted: 11/04/2010] [Indexed: 11/21/2022] Open
Abstract
Background The combined effects of multiple density-dependent, regulatory processes may have an important impact on the growth and stability of a population. In a malaria model system, it has been shown that the progression of Plasmodium berghei through Anopheles stephensi and the survival of the mosquito both depend non-linearly on parasite density. These processes regulating the development of the malaria parasite within the mosquito may influence the success of transmission-blocking interventions (TBIs) currently under development. Methods An individual-based stochastic mathematical model is used to investigate the combined impact of these multiple regulatory processes and examine how TBIs, which target different parasite life-stages within the mosquito, may influence overall parasite transmission. Results The best parasite molecular targets will vary between different epidemiological settings. Interventions that reduce ookinete density beneath a threshold level are likely to have auxiliary benefits, as transmission would be further reduced by density-dependent processes that restrict sporogonic development at low parasite densities. TBIs which reduce parasite density but fail to clear the parasite could cause a modest increase in transmission by increasing the number of infectious bites made by a mosquito during its lifetime whilst failing to sufficiently reduce its infectivity. Interventions with a higher variance in efficacy will therefore tend to cause a greater reduction in overall transmission than a TBI with a more uniform effectiveness. Care should be taken when interpreting these results as parasite intensity values in natural parasite-vector combinations of human malaria are likely to be significantly lower than those in this model system. Conclusions A greater understanding of the development of the malaria parasite within the mosquito is required to fully evaluate the impact of TBIs. If parasite-induced vector mortality influenced the population dynamics of Plasmodium species infecting humans in malaria endemic regions, it would be important to quantify the variability and duration of TBI efficacy to ensure that community benefits of control measures are not overestimated.
Collapse
Affiliation(s)
- Thomas S Churcher
- Department of Infectious Disease Epidemiology, School of Public Health, Faculty of Medicine, Imperial College London, UK.
| | | | | | | | | | | |
Collapse
|
41
|
Modelling malaria population structure and its implications for control. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 673:112-26. [PMID: 20632533 DOI: 10.1007/978-1-4419-6064-1_8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
Abstract
Mathematical models of malaria transmission have been used to inform the design of malaria control programs since the mid 20th century, and many of these models have provided useful insights into the complexity of the disease. Among developing countries, however and particularly in sub-Saharan Africa, malaria remains a major cause of morbidity and mortality. One of the main difficulties in controlling the most virulent human malaria parasite, Plasmodium falciparum, is its genetic diversity, which confounds attempts to design an effective vaccine. The population structure of P. falciparum remains poorly understood but plays a key role in determining epidemiological patterns of disease and the development of immunity. We discuss the seminal model of malaria transmission developed by Ross and MacDonald, and the modifications that have been made since to include more realism. We show that age profiles of disease and serological data support a theoretical model in which the parasite population is diverse and structured into several antigenic types and highlight the implications of this structure for controlling malaria. Lastly, we discuss the current sequence data on parasite antigen genes that are important for the aquisition of immunity, and the results of a new analysis of P. falciparum population structure at the genomic level.
Collapse
|
42
|
Safety and immunogenicity of a recombinant nonglycosylated erythrocyte binding antigen 175 Region II malaria vaccine in healthy adults living in an area where malaria is not endemic. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2010; 17:1552-9. [PMID: 20702657 DOI: 10.1128/cvi.00082-10] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Erythrocyte binding antigen region II (EBA-175) is a conserved antigen of Plasmodium falciparum that is involved in binding of the parasite to the host's erythrocytes. We evaluated the safety and immunogenicity of a recombinant EBA-175 vaccine with aluminum phosphate adjuvant in healthy young adults living in the United States. Eighteen subjects/group received ascending doses (5, 20, 80, or 160 μg) of the vaccine at 0, 1, and 6 months; 8 subjects received placebo. Most of the injection site and systemic reactions were mild to moderate in intensity. After 2 or 3 doses of the vaccine at any concentration, antibody levels measured by enzyme-linked immunosorbent assay were significantly higher than those for the placebo group. Sera from subjects who received 3 doses of the vaccine at any concentration inhibited the growth of erythrocyte-stage P. falciparum at low levels compared to sera from placebo recipients or preimmune sera. In conclusion, the EBA-175 vaccine with adjuvant was safe and immunogenic in malaria-naïve subjects.
Collapse
|
43
|
Hirai M, Mori T. Fertilization is a novel attacking site for the transmission blocking of malaria parasites. Acta Trop 2010; 114:157-61. [PMID: 19665985 DOI: 10.1016/j.actatropica.2009.08.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2009] [Revised: 07/21/2009] [Accepted: 08/04/2009] [Indexed: 01/20/2023]
Abstract
Malaria parasites perform sexual reproduction in mosquitoes where a pair of gametes fertilizes and differentiates into zygotes, and a single zygote produces several thousands of progeny infectious to next vertebrates. Although the parasite fertilization step has been considered as Achilles' heel of parasite life cycle and thus a critical target for blocking malaria transmission in the mosquito, its molecular mechanisms are largely unknown. Previously, we identified that GENERATIVE CELL SPECIFIC 1 (GCS1) is a reproduction factor in angiosperm. Subsequently, it was found that rodent malaria parasite, Plasmodium berghei and green algae, Chlamydomonas reinhardtii possess GCS1 homologues which also play essential roles in gamete interaction. Moreover, intensive database mining revealed that GCS1-like gene homologues exist in the genomes of various organisms. Thus, it appears that GCS1 is an ancient and highly conserved molecule functioning at gamete interaction. In this mini-review, we describe the mechanisms of gametogenesis and fertilization in malaria parasites, comparing with other eukaryotic reproduction, and also speculate GCS1 functions in gamete interaction. We discuss the possibility of whether malaria GCS1 is a novel type of transmission blocking vaccine, by which anti-malaria GCS1 antibody may halt parasite fertilization and subsequent developments in the mosquitoes.
Collapse
Affiliation(s)
- Makoto Hirai
- Division of Medical Zoology, Department of Infection and Immunity, Jichi Medical University, School of Medicine, Shimotsuke City, Tochigi, Japan.
| | | |
Collapse
|
44
|
Teboh-Ewungkem MI, Yuster T. A within-vector mathematical model of Plasmodium falciparum and implications of incomplete fertilization on optimal gametocyte sex ratio. J Theor Biol 2010; 264:273-86. [PMID: 20122943 DOI: 10.1016/j.jtbi.2009.12.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2009] [Revised: 12/11/2009] [Accepted: 12/14/2009] [Indexed: 10/19/2022]
Abstract
A mathematical model that simulates the within-vector dynamics of Plasmodium falciparum in an Anopheles mosquito is developed, based on experimental data. The model takes a mosquito's blood meal as input and computes the salivary gland sporozoite load as the final output, a probable measure of mosquito infectivity. Computational model results are consistent with observed results in nature. Sensitivity analysis of the model parameters suggests that reducing the gametocyte density in the blood meal most significantly lowers sporozoite load in the salivary glands and hence mosquito infectivity, and is thus an attractive target for malaria control. The model is used to investigate the implication of incomplete fertilization on optimal gametocyte sex ratio. For a single strain, the transition from complete fertilization to increasingly incomplete fertilization shifts that ratio from 1 to N, where N is the number of viable male gametes produced by a single male gametocyte, towards 1 to 1, which is demonstrated to be the limiting ratio analytically. This ratio is then shown to be an evolutionarily stable strategy as well in the limiting case.
Collapse
|
45
|
Li F, Patra KP, Yowell CA, Dame JB, Chin K, Vinetz JM. Apical surface expression of aspartic protease Plasmepsin 4, a potential transmission-blocking target of the plasmodium ookinete. J Biol Chem 2010; 285:8076-83. [PMID: 20056606 DOI: 10.1074/jbc.m109.063388] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
To invade its definitive host, the mosquito, the malaria parasite must cross the midgut peritrophic matrix that is composed of chitin cross-linked by chitin-binding proteins and then develop into an oocyst on the midgut basal lamina. Previous evidence indicates that Plasmodium ookinete-secreted chitinase is important in midgut invasion. The mechanistic role of other ookinete-secreted enzymes in midgut invasion has not been previously examined. De novo mass spectrometry sequencing of a protein obtained by benzamidine affinity column of Plasmodium gallinaceum ookinete axenic culture supernatant demonstrated the presence of an ookinete-secreted plasmepsin, an aspartic protease previously only known to be present in the digestive vacuole of asexual stage malaria parasites. This plasmepsin, the ortholog of Plasmodium falciparum plasmepsin 4, was designated PgPM4. PgPM4 and PgCHT2 (the P. gallinaceum ortholog of P. falciparum chitinase PfCHT1) are both localized on the ookinete apical surface, and both are present in micronemes. Aspartic protease inhibitors (peptidomimetic and natural product), calpain inhibitors, and anti-PgPM4 monoclonal antibodies significantly reduced parasite infectivity for mosquitoes. These results suggest that plasmepsin 4, previously known only to function in the digestive vacuole of asexual blood stage Plasmodium, plays a role in how the ookinete interacts with the mosquito midgut interactions as it becomes an oocyst. These data are the first to delineate a role for an aspartic protease in mediating Plasmodium invasion of the mosquito and demonstrate the potential for plasmepsin 4 as a malaria transmission-blocking vaccine target.
Collapse
Affiliation(s)
- Fengwu Li
- Division of Infectious Diseases, Department of Medicine, University of California San Diego School of Medicine, La Jolla, California 92093, USA
| | | | | | | | | | | |
Collapse
|
46
|
|
47
|
Teboh-Ewungkem MI, Podder CN, Gumel AB. Mathematical study of the role of gametocytes and an imperfect vaccine on malaria transmission dynamics. Bull Math Biol 2009; 72:63-93. [PMID: 19568725 DOI: 10.1007/s11538-009-9437-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2009] [Accepted: 06/02/2009] [Indexed: 11/27/2022]
Abstract
A mathematical model is developed to assess the role of gametocytes (the infectious sexual stage of the malaria parasite) in malaria transmission dynamics in a community. The model is rigorously analysed to gain insights into its dynamical features. It is shown that, in the absence of disease-induced mortality, the model has a globally-asymptotically stable disease-free equilibrium whenever a certain epidemiological threshold, known as the basic reproduction number (denoted by R(0)), is less than unity. Further, it has a unique endemic equilibrium if R(0) > 1. The model is extended to incorporate an imperfect vaccine with some assumed therapeutic characteristics. Theoretical analyses of the model with vaccination show that an imperfect malaria vaccine could have negative or positive impact (in reducing disease burden) depending on whether or not a certain threshold (denoted by nabla) is less than unity. Numerical simulations of the vaccination model show that such an imperfect anti-malaria vaccine (with a modest efficacy and coverage rate) can lead to effective disease control if the reproduction threshold (denoted by R(vac)) of the disease is reasonably small. On the other hand, the disease cannot be effectively controlled using such a vaccine if R(vac) is high. Finally, it is shown that the average number of days spent in the class of infectious individuals with higher level of gametocyte is critically important to the malaria burden in the community.
Collapse
|
48
|
Gholizadeh S, Djadid ND, Basseri HR, Zakeri S, Ladoni H. Analysis of von Willebrand factor A domain-related protein (WARP) polymorphism in temperate and tropical Plasmodium vivax field isolates. Malar J 2009; 8:137. [PMID: 19549316 PMCID: PMC2709902 DOI: 10.1186/1475-2875-8-137] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2009] [Accepted: 06/23/2009] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The identification of key molecules is crucial for designing transmission-blocking vaccines (TBVs), among those ookinete micronemal proteins are candidate as a general class of malaria transmission-blocking targets. Here, the sequence analysis of an extra-cellular malaria protein expressed in ookinetes, named von Willebrand factor A domain-related protein (WARP), is reported in 91 Plasmodium vivax isolates circulating in different regions of Iran. METHODS Clinical isolates were collected from north temperate and southern tropical regions in Iran. Primers have been designed based on P. vivax sequence (ctg_6991) which amplified a fragment of about 1044 bp with no size variation. Direct sequencing of PCR products was used to determine polymorphism and further bioinformatics analysis in P. vivax sexual stage antigen, pvwarp. RESULTS Amplified pvwarp gene showed 886 bp in size, with no intron. BLAST analysis showed a similarity of 98-100% to P. vivax Sal-I strain; however, Iranian isolates had 2 bp mismatches in 247 and 531 positions that were non-synonymous substitution [T (ACT) to A (GCT) and R (AGA) to S (AGT)] in comparison with the Sal-I sequence. CONCLUSION This study presents the first large-scale survey on pvwarp polymorphism in the world, which provides baseline data for developing WARP-based TBV against both temperate and tropical P. vivax isolates.
Collapse
Affiliation(s)
- Saber Gholizadeh
- Biotechnology Research Center (BRC), Pasteur Institute of Iran (PII), Tehran, Iran.
| | | | | | | | | |
Collapse
|
49
|
Sutherland CJ. Surface antigens of Plasmodium falciparum gametocytes--a new class of transmission-blocking vaccine targets? Mol Biochem Parasitol 2009; 166:93-8. [PMID: 19450726 DOI: 10.1016/j.molbiopara.2009.03.007] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2008] [Revised: 03/07/2009] [Accepted: 03/10/2009] [Indexed: 10/21/2022]
Abstract
The re-establishment of elimination and eradication on the malaria control agenda has led to calls for renewed effort in the development of parasite transmission-blocking interventions. Vaccines are ideally suited to this task, but progress towards an anti-gamete transmission-blocking vaccine, designed to act on parasites in blood-fed mosquitoes, has been slow. Recent work has confirmed that the surface of the gametocyte-infected erythrocyte presents antigens to the host immune system, and elicits specific humoral immune responses to these antigens, termed gametocyte surface antigens (GSAs). Likely candidate molecules, including antigens encoded by sub-telomeric multi-gene families, are discussed, and a hypothetical group of parasite molecules involved in spatial and temporal signal transduction in the human host is proposed, the tropins and circadins. The next steps for development of anti-gametocyte transmission-blocking vaccines for P. falciparum and the other human malaria species are considered.
Collapse
Affiliation(s)
- Colin J Sutherland
- HPA Malaria Reference Laboratory, Department of Infectious & Tropical Diseases, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom.
| |
Collapse
|
50
|
Simon N, Scholz SM, Moreira CK, Templeton TJ, Kuehn A, Dude MA, Pradel G. Sexual stage adhesion proteins form multi-protein complexes in the malaria parasite Plasmodium falciparum. J Biol Chem 2009; 284:14537-46. [PMID: 19304662 DOI: 10.1074/jbc.m808472200] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The sexual phase of the malaria parasite Plasmodium falciparum is accompanied by the coordinated expression of stage-specific adhesive proteins. Among these are six secreted proteins with multiple adhesion domains, termed P. falciparum LCCL domain-containing protein (PfCCp) proteins, which are expressed in the parasitophorous vacuole of the differentiating gametocytes and which are later associated with macrogametes. Although the majority of the PfCCp proteins are implicated in parasite development in the mosquito vector, their functions remain unknown. In the present study we investigated the molecular interactions between the PfCCp proteins during gametocyte development and emergence. Using five different gene-disruptant parasite lines, we show that the lack of one PfCCp protein leads to the loss of other PfCCp family members. Co-immunoprecipitation assays on gametocyte lysates revealed formation of complexes involving all PfCCp proteins, and affinity chromatography co-elution binding assays with recombinant PfCCp domains further indicated direct binding between distinct adhesion domains. PfCCp-coated latex beads bind to newly formed macrogametes but not to gametocytes or older macrogametes 6 or 24 h post-activation. In view of these data, we propose that the PfCCp proteins form multi-protein complexes that are exposed during gametogenesis, thereby mediating cell contacts of macrogametes.
Collapse
Affiliation(s)
- Nina Simon
- Research Center for Infectious Diseases, University of Würzburg, Röntgenring 11, 97070 Würzburg, Germany
| | | | | | | | | | | | | |
Collapse
|