247 results on '"Julian C Rayner"'
Search Results
2. The Dantu blood group prevents parasite growth in vivo: Evidence from a controlled human malaria infection study
- Author
-
Silvia N Kariuki, Alexander W Macharia, Johnstone Makale, Wilfred Nyamu, Stephen L Hoffman, Melissa C Kapulu, Philip Bejon, Julian C Rayner, Thomas N Williams, and On behalf of for the CHMI-SIKA Study Team
- Subjects
malaria ,challenge study ,host genetics ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Background: The long co-evolution of Homo sapiens and Plasmodium falciparum has resulted in the selection of numerous human genetic variants that confer an advantage against severe malaria and death. One such variant is the Dantu blood group antigen, which is associated with 74% protection against severe and complicated P. falciparum malaria infections in homozygous individuals, similar to that provided by the sickle haemoglobin allele (HbS). Recent in vitro studies suggest that Dantu exerts this protection by increasing the surface tension of red blood cells, thereby impeding the ability of P. falciparum merozoites to invade them and reducing parasite multiplication. However, no studies have yet explored this hypothesis in vivo. Methods: We investigated the effect of Dantu on early phase P. falciparum (Pf) infections in a controlled human malaria infection (CHMI) study. 141 sickle-negative Kenyan adults were inoculated with 3.2 × 103 aseptic, purified, cryopreserved Pf sporozoites (PfSPZ Challenge) then monitored for blood-stage parasitaemia for 21 days by quantitative polymerase chain reaction (qPCR)analysis of the 18S ribosomal RNA P. falciparum gene. The primary endpoint was blood-stage P. falciparum parasitaemia of ≥500/μl while the secondary endpoint was the receipt of antimalarial treatment in the presence of parasitaemia of any density. On study completion, all participants were genotyped both for Dantu and for four other polymorphisms that are associated with protection against severe falciparum malaria: α+-thalassaemia, blood group O, G6PD deficiency, and the rs4951074 allele in the red cell calcium transporter ATP2B4. Results: The primary endpoint was reached in 25/111 (22.5%) non-Dantu subjects in comparison to 0/27 (0%) Dantu heterozygotes and 0/3 (0.0%) Dantu homozygotes (p=0.01). Similarly, 49/111 (44.1%) non-Dantu subjects reached the secondary endpoint in comparison to only 7/27 (25.9%) and 0/3 (0.0%) Dantu heterozygotes and homozygotes, respectively (p=0.021). No significant impacts on either outcome were seen for any of the other genetic variants under study. Conclusions: This study reveals, for the first time, that the Dantu blood group is associated with high-level protection against early, non-clinical, P. falciparum malaria infections in vivo. Learning more about the mechanisms involved could potentially lead to new approaches to the prevention or treatment of the disease. Our study illustrates the power of CHMI with PfSPZ Challenge for directly testing the protective impact of genotypes previously identified using other methods. Funding: The Kenya CHMI study was supported by an award from Wellcome (grant number 107499). SK was supported by a Training Fellowship (216444/Z/19/Z), TNW by a Senior Research Fellowship (202800/Z/16/Z), JCR by an Investigator Award (220266/Z/20/Z), and core support to the KEMRI-Wellcome Trust Research Programme in Kilifi, Kenya (203077), all from Wellcome. The funders had no role in study design, data collection and interpretation, or the decision to submit the work for publication. For the purpose of Open Access, the authors have applied a CC BY public copyright license to any Author Accepted Manuscript version arising from this submission. Clinical trial number: NCT02739763
- Published
- 2023
- Full Text
- View/download PDF
3. Pf7: an open dataset of Plasmodium falciparum genome variation in 20,000 worldwide samples [version 1; peer review: 2 approved]
- Author
-
Mohamed Hassan Abdelraheem, Sonia Goncalves, Lemu Golassa, Tim Anderson, Desmond Omane Acheampong, Enoch Aninagyei, Ifeyinwa Aniebo, Patrick O Ansah, Felix Ansah, Gordon A Awandare, Paulo Arnaldo, Maciej F Boni, Gwladys I Bertin, Peter C Bull, Marielle Bouyou-Akotet, Keobouphaphone Chindavongsa, Edwin Kamau, Huch Cheah, Claire Kamaliddin, Vladimir Corredor, David J Conway, Nicholas Day, Abibatou Konaté, Erin Courtier, Theerarat Kochakarn, Arjen Dondorp, Abdoulaye Djimde, Diego F Echeverry, Seydou Doumbia, Mara Lawniczak, Pharath Lim, Sonia Maria Mauricio Enosse, Oumou Maïga-Ascofaré, Thomas G Egwang, Aung Myint Thu, Mark Fleharty, Jutta Marfurt, Caterina A Fanello, Mark Fukuda, Victor Mobegi, Matthew Forbes, Sara Anne Healy, G L Abby Harrison, Anastasia Hernandez-Koutoucheva, Jason A Hendry, Ivo Mueller, Francis Hombhanje, Harald Noedl, Catherine A Hill, Thuy-Nhien Nguyen, Mazza Hussein, Amanda Hott, Rintis Noviyanti, Scott A Jackson, Abraham Oduro, Deus Ishengoma, Harold Ocholla, Julia Jeans, Jean-Bosco Ouedraogo, Chris G Jacob, Drissa S Konate, Jon Keatley, Francois Nosten, Kolapo Oyebola, Myat P Kyaw, Aminatou Kone, Norbert Peshu, Samuel K Lee, Dennis Kyle, Kovana M Loua, Milijaona Randrianarivelojosia, Martha Lemnge, Sasithon Pukrittayakamee, Richard James Maude, Pascal Ringwald, Celine I Mandara, Abdelrahim Osman Mohamed, Toshihiro Mita, Jaqui Montgomery, Julian C Rayner, David Saunders, Olugbenga A Mokuolu, Lastenia Ruiz, Kathryn Murie, Peter Siba, Collins Misita Morang’a, Alex Shayo, Tuyen Nguyen Thi Kim, Thang Ngo Duc, Vincent Ntui-Njock Ntui, Colin Sutherland, Hong Nguyen Van, Xin-zhuan Su, Marie A Onyamboko, Livingstone Tavul, Irene Omedo, Richard Pearson, Antoinette Tshefu, Wellington Aghoghovwia Oyibo, Vandana Thathy, Chris Drakeley, Huynh Hong Quang, Joseph Vinetz, Christopher V Plowe, Federica Verra, Eduard Rovira-Vallbona, Jason Wendler, Anna Rosanas-Urgell, Teun Bousema, Thuy Nguyen, Mahamadou S. Sissoko, Valentin Ruano-Rubio, Alexis Nzila, Shannon Takala-Harrison, Christen Smith, William Yavo, Ngo Viet Thanh, Arthur Talman, Georgia Whitton, Mahamoudou Toure, Rob W van der Pluijm, Sarah Auburn, Antoine Claessens, Mahamadou Diakite, Kesinee Chotivanich, Mehul Dhorda, Olivo Miotto, Mallika Imwong, Mayfong Mayxay, Alfred Amambua-Ngwa, Philip Bejon, Elizabeth Ashley, Alyssa Barry, Rick M. Fairhurst, Ye Htut, Tran Tinh Hien, Kimberly J Johnson, Dominic P Kwiatkowski, Umberto D'Alessandro, Chanthap Lon, Paul N Newton, Aung P Phyo, Ric N Price, Victoria J Simpson, Kevin Marsh, Nicholas J White, Thomas E Wellems, Lynette Isabella Ochola-Oyier, Mozam Ali, Ambroise Ahouidi, Jacob Almagro-Garcia, Ben Andagalu, Lucas Amenga-Etego, Voahangy Andrianaranjaka, Tobias Apinjoh, Vito Baraka, Hampate Ba, Steffen Borrmann, Oralee Branch, Thanat Chookajorn, Souleymane Dama, Chanaki Amaratunga, Alister Craig, Brigitte Denis, Eleanor Drury, Christiane Dolecek, Patrick Duffy, Berhanu Erko, Abdul Faiz, Muzamil Mahdi Abdel Hamid, Anita Ghansah, and Dionicia Gamboa
- Subjects
malaria ,plasmodium falciparum ,genomics ,data resource ,genomic epidemiology ,eng ,Medicine ,Science - Abstract
We describe the MalariaGEN Pf7 data resource, the seventh release of Plasmodium falciparum genome variation data from the MalariaGEN network. It comprises over 20,000 samples from 82 partner studies in 33 countries, including several malaria endemic regions that were previously underrepresented. For the first time we include dried blood spot samples that were sequenced after selective whole genome amplification, necessitating new methods to genotype copy number variations. We identify a large number of newly emerging crt mutations in parts of Southeast Asia, and show examples of heterogeneities in patterns of drug resistance within Africa and within the Indian subcontinent. We describe the profile of variations in the C-terminal of the csp gene and relate this to the sequence used in the RTS,S and R21 malaria vaccines. Pf7 provides high-quality data on genotype calls for 6 million SNPs and short indels, analysis of large deletions that cause failure of rapid diagnostic tests, and systematic characterisation of six major drug resistance loci, all of which can be freely downloaded from the MalariaGEN website.
- Published
- 2023
- Full Text
- View/download PDF
4. Resolving drug selection and migration in an inbred South American Plasmodium falciparum population with identity-by-descent analysis.
- Author
-
Manuela Carrasquilla, Angela M Early, Aimee R Taylor, Angélica Knudson Ospina, Diego F Echeverry, Timothy J C Anderson, Elvira Mancilla, Samanda Aponte, Pablo Cárdenas, Caroline O Buckee, Julian C Rayner, Fabián E Sáenz, Daniel E Neafsey, and Vladimir Corredor
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
The human malaria parasite Plasmodium falciparum is globally widespread, but its prevalence varies significantly between and even within countries. Most population genetic studies in P. falciparum focus on regions of high transmission where parasite populations are large and genetically diverse, such as sub-Saharan Africa. Understanding population dynamics in low transmission settings, however, is of particular importance as these are often where drug resistance first evolves. Here, we use the Pacific Coast of Colombia and Ecuador as a model for understanding the population structure and evolution of Plasmodium parasites in small populations harboring less genetic diversity. The combination of low transmission and a high proportion of monoclonal infections means there are few outcrossing events and clonal lineages persist for long periods of time. Yet despite this, the population is evolutionarily labile and has successfully adapted to changes in drug regime. Using newly sequenced whole genomes, we measure relatedness between 166 parasites, calculated as identity by descent (IBD), and find 17 distinct but highly related clonal lineages, six of which have persisted in the region for at least a decade. This inbred population structure is captured in more detail with IBD than with other common population structure analyses like PCA, ADMIXTURE, and distance-based trees. We additionally use patterns of intra-chromosomal IBD and an analysis of haplotypic variation to explore past selection events in the region. Two genes associated with chloroquine resistance, crt and aat1, show evidence of hard selective sweeps, while selection appears soft and/or incomplete at three other key resistance loci (dhps, mdr1, and dhfr). Overall, this work highlights the strength of IBD analyses for studying parasite population structure and resistance evolution in regions of low transmission, and emphasizes that drug resistance can evolve and spread in small populations, as will occur in any region nearing malaria elimination.
- Published
- 2022
- Full Text
- View/download PDF
5. Longitudinal IgG antibody responses to Plasmodium vivax blood-stage antigens during and after acute vivax malaria in individuals living in the Brazilian Amazon.
- Author
-
Tenzin Tashi, Aditi Upadhye, Prasun Kundu, Chunxiang Wu, Sébastien Menant, Roberta Reis Soares, Marcelo U Ferreira, Rhea J Longley, Ivo Mueller, Quyen Q Hoang, Wai-Hong Tham, Julian C Rayner, Kézia Kg Scopel, Josué C Lima-Junior, and Tuan M Tran
- Subjects
Arctic medicine. Tropical medicine ,RC955-962 ,Public aspects of medicine ,RA1-1270 - Abstract
BackgroundTo make progress towards malaria elimination, a highly effective vaccine targeting Plasmodium vivax is urgently needed. Evaluating the kinetics of natural antibody responses to vaccine candidate antigens after acute vivax malaria can inform the design of serological markers of exposure and vaccines.Methodology/principal findingsThe responses of IgG antibodies to 9 P. vivax vaccine candidate antigens were evaluated in longitudinal serum samples from Brazilian individuals collected at the time of acute vivax malaria and 30, 60, and 180 days afterwards. Antigen-specific IgG correlations, seroprevalence, and half-lives were determined for each antigen using the longitudinal data. Antibody reactivities against Pv41 and PVX_081550 strongly correlated with each other at each of the four time points. The analysis identified robust responses in terms of magnitude and seroprevalence against Pv41 and PvGAMA at 30 and 60 days. Among the 8 P. vivax antigens demonstrating >50% seropositivity across all individuals, antibodies specific to PVX_081550 had the longest half-life (100 days; 95% CI, 83-130 days), followed by PvRBP2b (91 days; 95% CI, 76-110 days) and Pv12 (82 days; 95% CI, 64-110 days).Conclusion/significanceThis study provides an in-depth assessment of the kinetics of antibody responses to key vaccine candidate antigens in Brazilians with acute vivax malaria. Follow-up studies are needed to determine whether the longer-lived antibody responses induced by natural infection are effective in controlling blood-stage infection and mediating clinical protection.
- Published
- 2022
- Full Text
- View/download PDF
6. An open dataset of Plasmodium vivax genome variation in 1,895 worldwide samples [version 1; peer review: 2 approved]
- Author
-
Sonia Goncalves, Lemu Golassa, Wasif Khan, Sisay Alemu, Mohammad Shafiul Alam, Pharath Lim, Elizabeth Ashley, Nicholas M Anstey, Bridget E Barber, Jutta Marfurt, Ashenafi Assefa, Dhelio Batista Pereira, Alyssa Barry, Nguyen Hoang Chau, Jun Cao, Fe Espino, Cindy Chu, Ivo Mueller, María Fernanda Villegas, Rick Fairhurst, Thuy-Nhien Nguyen, Yaghoob Hamedi, Matthew J Grigg, Rintis Noviyanti, Ye Htut, Tran Tinh Hien, Nadira Karunaweera, Kimberly J Johnson, Dominic P Kwiatkowski, Srivicha Krudsood, Francois Nosten, Benedikt Ley, Marcus Lacerda, Alejandro Llanos-Cuentas, Yaobao Liu, Tatiana Lopera-Mesa, Milijaona Randrianarivelojosia, Chanthap Lon, Sasithon Pukrittayakamee, Rezika Mohammed, Pascal Michon, Paul N Newton, Chayadol Namaik-larp, Richard D Pearson, Julian C Rayner, Zuleima Pava, Aung P Phyo, Beyene Petros, Awab Ghulam Rahim, Ric N Price, Sasha V Siegel, Angela Rumaseb, Kamala Thriemer, Victoria J Simpson, Marcelo Urbano Ferreira, Alberto Tobon-Castano, Sonam Wangchuk, Ivan D Vélez, Nicholas J White, Thomas E Wellems, Maria F Yasnot, Arjen M. Dondorp, Timothy William, Daniel Yilma, Sarah Auburn, Hidayat Trimarsanto, Abraham Aseffa, Qi Gao, Roberto Amato, Voahangy Andrianaranjaka, Ishag Adam, Kesinee Chotivanich, Olivo Miotto, Chanaki Amaratunga, Eleanor Drury, Diego F. Echeverry, Berhanu Erko, and Abdul Faiz
- Subjects
malaria ,plasmodium vivax ,genomics ,data resource ,genomic epidemiology ,eng ,Medicine ,Science - Abstract
This report describes the MalariaGEN Pv4 dataset, a new release of curated genome variation data on 1,895 samples of Plasmodium vivax collected at 88 worldwide locations between 2001 and 2017. It includes 1,370 new samples contributed by MalariaGEN and VivaxGEN partner studies in addition to previously published samples from these and other sources. We provide genotype calls at over 4.5 million variable positions including over 3 million single nucleotide polymorphisms (SNPs), as well as short indels and tandem duplications. This enlarged dataset highlights major compartments of parasite population structure, with clear differentiation between Africa, Latin America, Oceania, Western Asia and different parts of Southeast Asia. Each sample has been classified for drug resistance to sulfadoxine, pyrimethamine and mefloquine based on known markers at the dhfr, dhps and mdr1 loci. The prevalence of all of these resistance markers was much higher in Southeast Asia and Oceania than elsewhere. This open resource of analysis-ready genome variation data from the MalariaGEN and VivaxGEN networks is driven by our collective goal to advance research into the complex biology of P. vivax and to accelerate genomic surveillance for malaria control and elimination.
- Published
- 2022
- Full Text
- View/download PDF
7. Using Plasmodium knowlesi as a model for screening Plasmodium vivax blood-stage malaria vaccine targets reveals new candidates.
- Author
-
Duncan N Ndegwa, Prasun Kundu, Jessica B Hostetler, Alejandro Marin-Menendez, Theo Sanderson, Kioko Mwikali, Lisa H Verzier, Rachael Coyle, Sophie Adjalley, and Julian C Rayner
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Plasmodium vivax is responsible for the majority of malaria cases outside Africa. Unlike P. falciparum, the P. vivax life-cycle includes a dormant liver stage, the hypnozoite, which can cause infection in the absence of mosquito transmission. An effective vaccine against P. vivax blood stages would limit symptoms and pathology from such recurrent infections, and therefore could play a critical role in the control of this species. Vaccine development in P. vivax, however, lags considerably behind P. falciparum, which has many identified targets with several having transitioned to Phase II testing. By contrast only one P. vivax blood-stage vaccine candidate based on the Duffy Binding Protein (PvDBP), has reached Phase Ia, in large part because the lack of a continuous in vitro culture system for P. vivax limits systematic screening of new candidates. We used the close phylogenetic relationship between P. vivax and P. knowlesi, for which an in vitro culture system in human erythrocytes exists, to test the scalability of systematic reverse vaccinology to identify and prioritise P. vivax blood-stage targets. A panel of P. vivax proteins predicted to function in erythrocyte invasion were expressed as full-length recombinant ectodomains in a mammalian expression system. Eight of these antigens were used to generate polyclonal antibodies, which were screened for their ability to recognize orthologous proteins in P. knowlesi. These antibodies were then tested for inhibition of growth and invasion of both wild type P. knowlesi and chimeric P. knowlesi lines modified using CRISPR/Cas9 to exchange P. knowlesi genes with their P. vivax orthologues. Candidates that induced antibodies that inhibited invasion to a similar level as PvDBP were identified, confirming the utility of P. knowlesi as a model for P. vivax vaccine development and prioritizing antigens for further follow up.
- Published
- 2021
- Full Text
- View/download PDF
8. Whole genome sequencing of Plasmodium vivax isolates reveals frequent sequence and structural polymorphisms in erythrocyte binding genes.
- Author
-
Anthony Ford, Daniel Kepple, Beka Raya Abagero, Jordan Connors, Richard Pearson, Sarah Auburn, Sisay Getachew, Colby Ford, Karthigayan Gunalan, Louis H Miller, Daniel A Janies, Julian C Rayner, Guiyun Yan, Delenasaw Yewhalaw, and Eugenia Lo
- Subjects
Arctic medicine. Tropical medicine ,RC955-962 ,Public aspects of medicine ,RA1-1270 - Abstract
Plasmodium vivax malaria is much less common in Africa than the rest of the world because the parasite relies primarily on the Duffy antigen/chemokine receptor (DARC) to invade human erythrocytes, and the majority of Africans are Duffy negative. Recently, there has been a dramatic increase in the reporting of P. vivax cases in Africa, with a high number of them being in Duffy negative individuals, potentially indicating P. vivax has evolved an alternative invasion mechanism that can overcome Duffy negativity. Here, we analyzed single nucleotide polymorphism (SNP) and copy number variation (CNV) in Whole Genome Sequence (WGS) data from 44 P. vivax samples isolated from symptomatic malaria patients in southwestern Ethiopia, where both Duffy positive and Duffy negative individuals are found. A total of 123,711 SNPs were detected, of which 22.7% were nonsynonymous and 77.3% were synonymous mutations. The largest number of SNPs were detected on chromosomes 9 (24,007 SNPs; 19.4% of total) and 10 (16,852 SNPs, 13.6% of total). There were particularly high levels of polymorphism in erythrocyte binding gene candidates including merozoite surface protein 1 (MSP1) and merozoite surface protein 3 (MSP3.5, MSP3.85 and MSP3.9). Two genes, MAEBL and MSP3.8 related to immunogenicity and erythrocyte binding function were detected with significant signals of positive selection. Variation in gene copy number was also concentrated in genes involved in host-parasite interactions, including the expansion of the Duffy binding protein gene (PvDBP) on chromosome 6 and MSP3.11 on chromosome 10. Based on the phylogeny constructed from the whole genome sequences, the expansion of these genes was an independent process among the P. vivax lineages in Ethiopia. We further inferred transmission patterns of P. vivax infections among study sites and showed various levels of gene flow at a small geographical scale. The genomic features of P. vivax provided baseline data for future comparison with those in Duffy-negative individuals and allowed us to develop a panel of informative Single Nucleotide Polymorphic markers diagnostic at a micro-geographical scale.
- Published
- 2020
- Full Text
- View/download PDF
9. An enhanced toolkit for the generation of knockout and marker-free fluorescent Plasmodium chabaudi [version 2; peer review: 2 approved]
- Author
-
Edward J Marr, Rachel M Milne, Burcu Anar, Gareth Girling, Frank Schwach, Jason P Mooney, Wiebke Nahrendorf, Philip J Spence, Deirdre Cunningham, David A Baker, Jean Langhorne, Julian C Rayner, Oliver Billker, Ellen S Bushell, and Joanne Thompson
- Subjects
Medicine ,Science - Abstract
The rodent parasite Plasmodium chabaudi is an important in vivo model of malaria. The ability to produce chronic infections makes it particularly useful for investigating the development of anti-Plasmodium immunity, as well as features associated with parasite virulence during both the acute and chronic phases of infection. P. chabaudi also undergoes asexual maturation (schizogony) and erythrocyte invasion in culture, so offers an experimentally-amenable in vivo to in vitro model for studying gene function and drug activity during parasite replication. To extend the usefulness of this model, we have further optimised transfection protocols and plasmids for P. chabaudi and generated stable, fluorescent lines that are free from drug-selectable marker genes. These mother-lines show the same infection dynamics as wild-type parasites throughout the lifecycle in mice and mosquitoes; furthermore, their virulence can be increased by serial blood passage and reset by mosquito transmission. We have also adapted the large-insert, linear PlasmoGEM vectors that have revolutionised the scale of experimental genetics in another rodent malaria parasite and used these to generate barcoded P. chabaudi gene-deletion and –tagging vectors for transfection in our fluorescent P. chabaudi mother-lines. This produces a tool-kit of P. chabaudi lines, vectors and transfection approaches that will be of broad utility to the research community.
- Published
- 2020
- Full Text
- View/download PDF
10. An enhanced toolkit for the generation of knockout and marker-free fluorescent Plasmodium chabaudi [version 1; peer review: 2 approved]
- Author
-
Edward J Marr, Rachel M Milne, Burcu Anar, Gareth Girling, Frank Schwach, Jason P Mooney, Wiebke Nahrendorf, Philip J Spence, Deirdre Cunningham, David A Baker, Jean Langhorne, Julian C Rayner, Oliver Billker, Ellen S Bushell, and Joanne Thompson
- Subjects
Medicine ,Science - Abstract
The rodent parasite Plasmodium chabaudi is an important in vivo model of malaria. The ability to produce chronic infections makes it particularly useful for investigating the development of anti-Plasmodium immunity, as well as features associated with parasite virulence during both the acute and chronic phases of infection. P. chabaudi also undergoes asexual maturation (schizogony) and erythrocyte invasion in culture, so offers an experimentally-amenable in vivo to in vitro model for studying gene function and drug activity during parasite replication. To extend the usefulness of this model, we have further optimised transfection protocols and plasmids for P. chabaudi and generated stable, fluorescent lines that are free from drug-selectable marker genes. These mother-lines show the same infection dynamics as wild-type parasites throughout the lifecycle in mice and mosquitoes; furthermore, their virulence can be increased by serial blood passage and reset by mosquito transmission. We have also adapted the large-insert, linear PlasmoGEM vectors that have revolutionised the scale of experimental genetics in another rodent malaria parasite and used these to generate barcoded P. chabaudi gene-deletion and –tagging vectors for transfection in our fluorescent P. chabaudi mother-lines. This produces a tool-kit of P. chabaudi lines, vectors and transfection approaches that will be of broad utility to the research community.
- Published
- 2020
- Full Text
- View/download PDF
11. Frequent expansion of Plasmodium vivax Duffy Binding Protein in Ethiopia and its epidemiological significance.
- Author
-
Eugenia Lo, Jessica B Hostetler, Delenasaw Yewhalaw, Richard D Pearson, Muzamil M A Hamid, Karthigayan Gunalan, Daniel Kepple, Anthony Ford, Daniel A Janies, Julian C Rayner, Louis H Miller, and Guiyun Yan
- Subjects
Arctic medicine. Tropical medicine ,RC955-962 ,Public aspects of medicine ,RA1-1270 - Abstract
Plasmodium vivax invasion of human erythrocytes depends on the Duffy Binding Protein (PvDBP) which interacts with the Duffy antigen. PvDBP copy number has been recently shown to vary between P. vivax isolates in Sub-Saharan Africa. However, the extent of PvDBP copy number variation, the type of PvDBP multiplications, as well as its significance across broad samples are still unclear. We determined the prevalence and type of PvDBP duplications, as well as PvDBP copy number variation among 178 Ethiopian P. vivax isolates using a PCR-based diagnostic method, a novel quantitative real-time PCR assay and whole genome sequencing. For the 145 symptomatic samples, PvDBP duplications were detected in 95 isolates, of which 81 had the Cambodian and 14 Malagasy-type PvDBP duplications. PvDBP varied from 1 to >4 copies. Isolates with multiple PvDBP copies were found to be higher in symptomatic than asymptomatic infections. For the 33 asymptomatic samples, PvDBP was detected with two copies in two of the isolates, and both were the Cambodian-type PvDBP duplication. PvDBP copy number in Duffy-negative heterozygotes was not significantly different from that in Duffy-positives, providing no support for the hypothesis that increased copy number is a specific association with Duffy-negativity, although the number of Duffy-negatives was small and further sampling is required to test this association thoroughly.
- Published
- 2019
- Full Text
- View/download PDF
12. Plasmodium knowlesi as a model system for characterising Plasmodium vivax drug resistance candidate genes.
- Author
-
Lisa H Verzier, Rachael Coyle, Shivani Singh, Theo Sanderson, and Julian C Rayner
- Subjects
Arctic medicine. Tropical medicine ,RC955-962 ,Public aspects of medicine ,RA1-1270 - Abstract
Plasmodium vivax causes the majority of malaria outside Africa, but is poorly understood at a cellular level partly due to technical difficulties in maintaining it in in vitro culture conditions. In the past decades, drug resistant P. vivax parasites have emerged, mainly in Southeast Asia, but while some molecular markers of resistance have been identified, none have so far been confirmed experimentally, which limits interpretation of the markers, and hence our ability to monitor and control the spread of resistance. Some of these potential markers have been identified through P. vivax genome-wide population genetic analyses, which highlighted genes under recent evolutionary selection in Southeast Asia, where chloroquine resistance is most prevalent. These genes could be involved in drug resistance, but no experimental proof currently exists to support this hypothesis. In this study, we used Plasmodium knowlesi, the most closely related species to P. vivax that can be cultured in human erythrocytes, as a model system to express P. vivax genes and test for their role in drug resistance. We adopted a strategy of episomal expression, and were able to express fourteen P. vivax genes, including two allelic variants of several hypothetical resistance genes. Their expression level and localisation were assessed, confirming cellular locations conjectured from orthologous species, and suggesting locations for several previously unlocalised proteins, including an apical location for PVX_101445. These findings establish P. knowlesi as a suitable model for P. vivax protein expression. We performed chloroquine and mefloquine drug assays, finding no significant differences in drug sensitivity: these results could be due to technical issues, or could indicate that these genes are not actually involved in drug resistance, despite being under positive selection pressure in Southeast Asia. These data confirm that in vitro P. knowlesi is a useful tool for studying P. vivax biology. Its close evolutionary relationship to P. vivax, high transfection efficiency, and the availability of markers for colocalisation, all make it a powerful model system. Our study is the first of its kind using P. knowlesi to study unknown P. vivax proteins and investigate drug resistance mechanisms.
- Published
- 2019
- Full Text
- View/download PDF
13. A forward genetic screen reveals a primary role for Plasmodium falciparum Reticulocyte Binding Protein Homologue 2a and 2b in determining alternative erythrocyte invasion pathways.
- Author
-
Susana Campino, Alejandro Marin-Menendez, Alison Kemp, Nadia Cross, Laura Drought, Thomas D Otto, Ernest Diez Benavente, Matt Ravenhall, Frank Schwach, Gareth Girling, Magnus Manske, Michel Theron, Kelda Gould, Eleanor Drury, Taane G Clark, Dominic P Kwiatkowski, Alena Pance, and Julian C Rayner
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Invasion of human erythrocytes is essential for Plasmodium falciparum parasite survival and pathogenesis, and is also a complex phenotype. While some later steps in invasion appear to be invariant and essential, the earlier steps of recognition are controlled by a series of redundant, and only partially understood, receptor-ligand interactions. Reverse genetic analysis of laboratory adapted strains has identified multiple genes that when deleted can alter invasion, but how the relative contributions of each gene translate to the phenotypes of clinical isolates is far from clear. We used a forward genetic approach to identify genes responsible for variable erythrocyte invasion by phenotyping the parents and progeny of previously generated experimental genetic crosses. Linkage analysis using whole genome sequencing data revealed a single major locus was responsible for the majority of phenotypic variation in two invasion pathways. This locus contained the PfRh2a and PfRh2b genes, members of one of the major invasion ligand gene families, but not widely thought to play such a prominent role in specifying invasion phenotypes. Variation in invasion pathways was linked to significant differences in PfRh2a and PfRh2b expression between parasite lines, and their role in specifying alternative invasion was confirmed by CRISPR-Cas9-mediated genome editing. Expansion of the analysis to a large set of clinical P. falciparum isolates revealed common deletions, suggesting that variation at this locus is a major cause of invasion phenotypic variation in the endemic setting. This work has implications for blood-stage vaccine development and will help inform the design and location of future large-scale studies of invasion in clinical isolates.
- Published
- 2018
- Full Text
- View/download PDF
14. RecQ helicases in the malaria parasite Plasmodium falciparum affect genome stability, gene expression patterns and DNA replication dynamics.
- Author
-
Antoine Claessens, Lynne M Harris, Slavica Stanojcic, Lia Chappell, Adam Stanton, Nada Kuk, Pamela Veneziano-Broccia, Yvon Sterkers, Julian C Rayner, and Catherine J Merrick
- Subjects
Genetics ,QH426-470 - Abstract
The malaria parasite Plasmodium falciparum has evolved an unusual genome structure. The majority of the genome is relatively stable, with mutation rates similar to most eukaryotic species. However, some regions are very unstable with high recombination rates, driving the generation of new immune evasion-associated var genes. The molecular factors controlling the inconsistent stability of this genome are not known. Here we studied the roles of the two putative RecQ helicases in P. falciparum, PfBLM and PfWRN. When PfWRN was knocked down, recombination rates increased four-fold, generating chromosomal abnormalities, a high rate of chimeric var genes and many microindels, particularly in known 'fragile sites'. This is the first identification of a gene involved in suppressing recombination and maintaining genome stability in Plasmodium. By contrast, no change in mutation rate appeared when the second RecQ helicase, PfBLM, was mutated. At the transcriptional level, however, both helicases evidently modulate the transcription of large cohorts of genes, with several hundred genes-including a large proportion of vars-showing deregulated expression in each RecQ mutant. Aberrant processing of stalled replication forks is a possible mechanism underlying elevated mutation rates and this was assessed by measuring DNA replication dynamics in the RecQ mutant lines. Replication forks moved slowly and stalled at elevated rates in both mutants, confirming that RecQ helicases are required for efficient DNA replication. Overall, this work identifies the Plasmodium RecQ helicases as major players in DNA replication, antigenic diversification and genome stability in the most lethal human malaria parasite, with important implications for genome evolution in this pathogen.
- Published
- 2018
- Full Text
- View/download PDF
15. Identification of highly-protective combinations of Plasmodium vivax recombinant proteins for vaccine development
- Author
-
Camila Tenorio França, Michael T White, Wen-Qiang He, Jessica B Hostetler, Jessica Brewster, Gabriel Frato, Indu Malhotra, Jakub Gruszczyk, Christele Huon, Enmoore Lin, Benson Kiniboro, Anjali Yadava, Peter Siba, Mary R Galinski, Julie Healer, Chetan Chitnis, Alan F Cowman, Eizo Takashima, Takafumi Tsuboi, Wai-Hong Tham, Rick M Fairhurst, Julian C Rayner, Christopher L King, and Ivo Mueller
- Subjects
Plasmodium vivax ,IgG antibody ,vaccine ,clinical malaria ,protection ,natural immunity ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
The study of antigenic targets of naturally-acquired immunity is essential to identify and prioritize antigens for further functional characterization. We measured total IgG antibodies to 38 P. vivax antigens, investigating their relationship with prospective risk of malaria in a cohort of 1–3 years old Papua New Guinean children. Using simulated annealing algorithms, the potential protective efficacy of antibodies to multiple antigen-combinations, and the antibody thresholds associated with protection were investigated for the first time. High antibody levels to multiple known and newly identified proteins were strongly associated with protection (IRR 0.44–0.74, p90%), EBP, DBPII, RBP1a, CyRPA, and PVX_081550 were most frequently identified; several of them requiring very low antibody levels to show a protective association. These data identify individual antigens that should be prioritized for further functional testing and establish a clear path to testing a multicomponent P. vivax vaccine.
- Published
- 2017
- Full Text
- View/download PDF
16. The exported chaperone Hsp70-x supports virulence functions for Plasmodium falciparum blood stage parasites.
- Author
-
Sarah C Charnaud, Matthew W A Dixon, Catherine Q Nie, Lia Chappell, Paul R Sanders, Thomas Nebl, Eric Hanssen, Matthew Berriman, Jo-Anne Chan, Adam J Blanch, James G Beeson, Julian C Rayner, Jude M Przyborski, Leann Tilley, Brendan S Crabb, and Paul R Gilson
- Subjects
Medicine ,Science - Abstract
Malaria is caused by five different Plasmodium spp. in humans each of which modifies the host erythrocyte to survive and replicate. The two main causes of malaria, P. falciparum and P. vivax, differ in their ability to cause severe disease, mainly due to differences in the cytoadhesion of infected erythrocytes (IE) in the microvasculature. Cytoadhesion of P. falciparum in the brain leads to a large number of deaths each year and is a consequence of exported parasite proteins, some of which modify the erythrocyte cytoskeleton while others such as PfEMP1 project onto the erythrocyte surface where they bind to endothelial cells. Here we investigate the effects of knocking out an exported Hsp70-type chaperone termed Hsp70-x that is present in P. falciparum but not P. vivax. Although the growth of Δhsp70-x parasites was unaffected, the export of PfEMP1 cytoadherence proteins was delayed and Δhsp70-x IE had reduced adhesion. The Δhsp70-x IE were also more rigid than wild-type controls indicating changes in the way the parasites modified their host erythrocyte. To investigate the cause of this, transcriptional and translational changes in exported and chaperone proteins were monitored and some changes were observed. We propose that PfHsp70-x is not essential for survival in vitro, but may be required for the efficient export and functioning of some P. falciparum exported proteins.
- Published
- 2017
- Full Text
- View/download PDF
17. Independent Origin and Global Distribution of Distinct Plasmodium vivax Duffy Binding Protein Gene Duplications.
- Author
-
Jessica B Hostetler, Eugenia Lo, Usheer Kanjee, Chanaki Amaratunga, Seila Suon, Sokunthea Sreng, Sivanna Mao, Delenasaw Yewhalaw, Anjali Mascarenhas, Dominic P Kwiatkowski, Marcelo U Ferreira, Pradipsinh K Rathod, Guiyun Yan, Rick M Fairhurst, Manoj T Duraisingh, and Julian C Rayner
- Subjects
Arctic medicine. Tropical medicine ,RC955-962 ,Public aspects of medicine ,RA1-1270 - Abstract
BACKGROUND:Plasmodium vivax causes the majority of malaria episodes outside Africa, but remains a relatively understudied pathogen. The pathology of P. vivax infection depends critically on the parasite's ability to recognize and invade human erythrocytes. This invasion process involves an interaction between P. vivax Duffy Binding Protein (PvDBP) in merozoites and the Duffy antigen receptor for chemokines (DARC) on the erythrocyte surface. Whole-genome sequencing of clinical isolates recently established that some P. vivax genomes contain two copies of the PvDBP gene. The frequency of this duplication is particularly high in Madagascar, where there is also evidence for P. vivax infection in DARC-negative individuals. The functional significance and global prevalence of this duplication, and whether there are other copy number variations at the PvDBP locus, is unknown. METHODOLOGY/PRINCIPAL FINDINGS:Using whole-genome sequencing and PCR to study the PvDBP locus in P. vivax clinical isolates, we found that PvDBP duplication is widespread in Cambodia. The boundaries of the Cambodian PvDBP duplication differ from those previously identified in Madagascar, meaning that current molecular assays were unable to detect it. The Cambodian PvDBP duplication did not associate with parasite density or DARC genotype, and ranged in prevalence from 20% to 38% over four annual transmission seasons in Cambodia. This duplication was also present in P. vivax isolates from Brazil and Ethiopia, but not India. CONCLUSIONS/SIGNIFICANCE:PvDBP duplications are much more widespread and complex than previously thought, and at least two distinct duplications are circulating globally. The same duplication boundaries were identified in parasites from three continents, and were found at high prevalence in human populations where DARC-negativity is essentially absent. It is therefore unlikely that PvDBP duplication is associated with infection of DARC-negative individuals, but functional tests will be required to confirm this hypothesis.
- Published
- 2016
- Full Text
- View/download PDF
18. An Antibody Screen of a Plasmodium vivax Antigen Library Identifies Novel Merozoite Proteins Associated with Clinical Protection.
- Author
-
Camila T França, Jessica B Hostetler, Sumana Sharma, Michael T White, Enmoore Lin, Benson Kiniboro, Andreea Waltmann, Andrew W Darcy, Connie S N Li Wai Suen, Peter Siba, Christopher L King, Julian C Rayner, Rick M Fairhurst, and Ivo Mueller
- Subjects
Arctic medicine. Tropical medicine ,RC955-962 ,Public aspects of medicine ,RA1-1270 - Abstract
BACKGROUND:Elimination of Plasmodium vivax malaria would be greatly facilitated by the development of an effective vaccine. A comprehensive and systematic characterization of antibodies to P. vivax antigens in exposed populations is useful in guiding rational vaccine design. METHODOLOGY/PRINCIPAL FINDINGS:In this study, we investigated antibodies to a large library of P. vivax entire ectodomain merozoite proteins in 2 Asia-Pacific populations, analysing the relationship of antibody levels with markers of current and cumulative malaria exposure, and socioeconomic and clinical indicators. 29 antigenic targets of natural immunity were identified. Of these, 12 highly-immunogenic proteins were strongly associated with age and thus cumulative lifetime exposure in Solomon Islanders (P
- Published
- 2016
- Full Text
- View/download PDF
19. A Stem Cell Strategy Identifies Glycophorin C as a Major Erythrocyte Receptor for the Rodent Malaria Parasite Plasmodium berghei.
- Author
-
Loukia Yiangou, Ruddy Montandon, Katarzyna Modrzynska, Barry Rosen, Wendy Bushell, Christine Hale, Oliver Billker, Julian C Rayner, and Alena Pance
- Subjects
Medicine ,Science - Abstract
The clinical complications of malaria are caused by the parasite expansion in the blood. Invasion of erythrocytes is a complex process that depends on multiple receptor-ligand interactions. Identification of host receptors is paramount for fighting the disease as it could reveal new intervention targets, but the enucleated nature of erythrocytes makes genetic approaches impossible and many receptors remain unknown. Host-parasite interactions evolve rapidly and are therefore likely to be species-specific. As a results, understanding of invasion receptors outside the major human pathogen Plasmodium falciparum is very limited. Here we use mouse embryonic stem cells (mESCs) that can be genetically engineered and differentiated into erythrocytes to identify receptors for the rodent malaria parasite Plasmodium berghei. Two proteins previously implicated in human malaria infection: glycophorin C (GYPC) and Band-3 (Slc4a1) were deleted in mESCs to generate stable cell lines, which were differentiated towards erythropoiesis. In vitro infection assays revealed that while deletion of Band-3 has no effect, absence of GYPC results in a dramatic decrease in invasion, demonstrating the crucial role of this protein for P. berghei infection. This stem cell approach offers the possibility of targeting genes that may be essential and therefore difficult to disrupt in whole organisms and has the potential to be applied to a variety of parasites in diverse host cell types.
- Published
- 2016
- Full Text
- View/download PDF
20. A Library of Plasmodium vivax Recombinant Merozoite Proteins Reveals New Vaccine Candidates and Protein-Protein Interactions.
- Author
-
Jessica B Hostetler, Sumana Sharma, S Josefin Bartholdson, Gavin J Wright, Rick M Fairhurst, and Julian C Rayner
- Subjects
Arctic medicine. Tropical medicine ,RC955-962 ,Public aspects of medicine ,RA1-1270 - Abstract
BACKGROUND:A vaccine targeting Plasmodium vivax will be an essential component of any comprehensive malaria elimination program, but major gaps in our understanding of P. vivax biology, including the protein-protein interactions that mediate merozoite invasion of reticulocytes, hinder the search for candidate antigens. Only one ligand-receptor interaction has been identified, that between P. vivax Duffy Binding Protein (PvDBP) and the erythrocyte Duffy Antigen Receptor for Chemokines (DARC), and strain-specific immune responses to PvDBP make it a complex vaccine target. To broaden the repertoire of potential P. vivax merozoite-stage vaccine targets, we exploited a recent breakthrough in expressing full-length ectodomains of Plasmodium proteins in a functionally-active form in mammalian cells and initiated a large-scale study of P. vivax merozoite proteins that are potentially involved in reticulocyte binding and invasion. METHODOLOGY/PRINCIPAL FINDINGS:We selected 39 P. vivax proteins that are predicted to localize to the merozoite surface or invasive secretory organelles, some of which show homology to P. falciparum vaccine candidates. Of these, we were able to express 37 full-length protein ectodomains in a mammalian expression system, which has been previously used to express P. falciparum invasion ligands such as PfRH5. To establish whether the expressed proteins were correctly folded, we assessed whether they were recognized by antibodies from Cambodian patients with acute vivax malaria. IgG from these samples showed at least a two-fold change in reactivity over naïve controls in 27 of 34 antigens tested, and the majority showed heat-labile IgG immunoreactivity, suggesting the presence of conformation-sensitive epitopes and native tertiary protein structures. Using a method specifically designed to detect low-affinity, extracellular protein-protein interactions, we confirmed a predicted interaction between P. vivax 6-cysteine proteins P12 and P41, further suggesting that the proteins are natively folded and functional. This screen also identified two novel protein-protein interactions, between P12 and PVX_110945, and between MSP3.10 and MSP7.1, the latter of which was confirmed by surface plasmon resonance. CONCLUSIONS/SIGNIFICANCE:We produced a new library of recombinant full-length P. vivax ectodomains, established that the majority of them contain tertiary structure, and used them to identify predicted and novel protein-protein interactions. As well as identifying new interactions for further biological studies, this library will be useful in identifying P. vivax proteins with vaccine potential, and studying P. vivax malaria pathogenesis and immunity. TRIAL REGISTRATION:ClinicalTrials.gov NCT00663546.
- Published
- 2015
- Full Text
- View/download PDF
21. Revealing the sequence and resulting cellular morphology of receptor-ligand interactions during Plasmodium falciparum invasion of erythrocytes.
- Author
-
Greta E Weiss, Paul R Gilson, Tana Taechalertpaisarn, Wai-Hong Tham, Nienke W M de Jong, Katherine L Harvey, Freya J I Fowkes, Paul N Barlow, Julian C Rayner, Gavin J Wright, Alan F Cowman, and Brendan S Crabb
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
During blood stage Plasmodium falciparum infection, merozoites invade uninfected erythrocytes via a complex, multistep process involving a series of distinct receptor-ligand binding events. Understanding each element in this process increases the potential to block the parasite's life cycle via drugs or vaccines. To investigate specific receptor-ligand interactions, they were systematically blocked using a combination of genetic deletion, enzymatic receptor cleavage and inhibition of binding via antibodies, peptides and small molecules, and the resulting temporal changes in invasion and morphological effects on erythrocytes were filmed using live cell imaging. Analysis of the videos have shown receptor-ligand interactions occur in the following sequence with the following cellular morphologies; 1) an early heparin-blockable interaction which weakly deforms the erythrocyte, 2) EBA and PfRh ligands which strongly deform the erythrocyte, a process dependant on the merozoite's actin-myosin motor, 3) a PfRh5-basigin binding step which results in a pore or opening between parasite and host through which it appears small molecules and possibly invasion components can flow and 4) an AMA1-RON2 interaction that mediates tight junction formation, which acts as an anchor point for internalization. In addition to enhancing general knowledge of apicomplexan biology, this work provides a rational basis to combine sequentially acting merozoite vaccine candidates in a single multi-receptor-blocking vaccine.
- Published
- 2015
- Full Text
- View/download PDF
22. Plasmodium knowlesi genome sequences from clinical isolates reveal extensive genomic dimorphism.
- Author
-
Miguel M Pinheiro, Md Atique Ahmed, Scott B Millar, Theo Sanderson, Thomas D Otto, Woon Chan Lu, Sanjeev Krishna, Julian C Rayner, and Janet Cox-Singh
- Subjects
Medicine ,Science - Abstract
Plasmodium knowlesi is a newly described zoonosis that causes malaria in the human population that can be severe and fatal. The study of P. knowlesi parasites from human clinical isolates is relatively new and, in order to obtain maximum information from patient sample collections, we explored the possibility of generating P. knowlesi genome sequences from archived clinical isolates. Our patient sample collection consisted of frozen whole blood samples that contained excessive human DNA contamination and, in that form, were not suitable for parasite genome sequencing. We developed a method to reduce the amount of human DNA in the thawed blood samples in preparation for high throughput parasite genome sequencing using Illumina HiSeq and MiSeq sequencing platforms. Seven of fifteen samples processed had sufficiently pure P. knowlesi DNA for whole genome sequencing. The reads were mapped to the P. knowlesi H strain reference genome and an average mapping of 90% was obtained. Genes with low coverage were removed leaving 4623 genes for subsequent analyses. Previously we identified a DNA sequence dimorphism on a small fragment of the P. knowlesi normocyte binding protein xa gene on chromosome 14. We used the genome data to assemble full-length Pknbpxa sequences and discovered that the dimorphism extended along the gene. An in-house algorithm was developed to detect SNP sites co-associating with the dimorphism. More than half of the P. knowlesi genome was dimorphic, involving genes on all chromosomes and suggesting that two distinct types of P. knowlesi infect the human population in Sarawak, Malaysian Borneo. We use P. knowlesi clinical samples to demonstrate that Plasmodium DNA from archived patient samples can produce high quality genome data. We show that analyses, of even small numbers of difficult clinical malaria isolates, can generate comprehensive genomic information that will improve our understanding of malaria parasite diversity and pathobiology.
- Published
- 2015
- Full Text
- View/download PDF
23. Generation of antigenic diversity in Plasmodium falciparum by structured rearrangement of Var genes during mitosis.
- Author
-
Antoine Claessens, William L Hamilton, Mihir Kekre, Thomas D Otto, Adnan Faizullabhoy, Julian C Rayner, and Dominic Kwiatkowski
- Subjects
Genetics ,QH426-470 - Abstract
The most polymorphic gene family in P. falciparum is the ∼60 var genes distributed across parasite chromosomes, both in the subtelomeres and in internal regions. They encode hypervariable surface proteins known as P. falciparum erythrocyte membrane protein 1 (PfEMP1) that are critical for pathogenesis and immune evasion in Plasmodium falciparum. How var gene sequence diversity is generated is not currently completely understood. To address this, we constructed large clone trees and performed whole genome sequence analysis to study the generation of novel var gene sequences in asexually replicating parasites. While single nucleotide polymorphisms (SNPs) were scattered across the genome, structural variants (deletions, duplications, translocations) were focused in and around var genes, with considerable variation in frequency between strains. Analysis of more than 100 recombination events involving var exon 1 revealed that the average nucleotide sequence identity of two recombining exons was only 63% (range: 52.7-72.4%) yet the crossovers were error-free and occurred in such a way that the resulting sequence was in frame and domain architecture was preserved. Var exon 1, which encodes the immunologically exposed part of the protein, recombined in up to 0.2% of infected erythrocytes in vitro per life cycle. The high rate of var exon 1 recombination indicates that millions of new antigenic structures could potentially be generated each day in a single infected individual. We propose a model whereby var gene sequence polymorphism is mainly generated during the asexual part of the life cycle.
- Published
- 2014
- Full Text
- View/download PDF
24. Phosphoinositide metabolism links cGMP-dependent protein kinase G to essential Ca²⁺ signals at key decision points in the life cycle of malaria parasites.
- Author
-
Mathieu Brochet, Mark O Collins, Terry K Smith, Eloise Thompson, Sarah Sebastian, Katrin Volkmann, Frank Schwach, Lia Chappell, Ana Rita Gomes, Matthew Berriman, Julian C Rayner, David A Baker, Jyoti Choudhary, and Oliver Billker
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Many critical events in the Plasmodium life cycle rely on the controlled release of Ca²⁺ from intracellular stores to activate stage-specific Ca²⁺-dependent protein kinases. Using the motility of Plasmodium berghei ookinetes as a signalling paradigm, we show that the cyclic guanosine monophosphate (cGMP)-dependent protein kinase, PKG, maintains the elevated level of cytosolic Ca²⁺ required for gliding motility. We find that the same PKG-dependent pathway operates upstream of the Ca²⁺ signals that mediate activation of P. berghei gametocytes in the mosquito and egress of Plasmodium falciparum merozoites from infected human erythrocytes. Perturbations of PKG signalling in gliding ookinetes have a marked impact on the phosphoproteome, with a significant enrichment of in vivo regulated sites in multiple pathways including vesicular trafficking and phosphoinositide metabolism. A global analysis of cellular phospholipids demonstrates that in gliding ookinetes PKG controls phosphoinositide biosynthesis, possibly through the subcellular localisation or activity of lipid kinases. Similarly, phosphoinositide metabolism links PKG to egress of P. falciparum merozoites, where inhibition of PKG blocks hydrolysis of phosphatidylinostitol (4,5)-bisphosphate. In the face of an increasing complexity of signalling through multiple Ca²⁺ effectors, PKG emerges as a unifying factor to control multiple cellular Ca²⁺ signals essential for malaria parasite development and transmission.
- Published
- 2014
- Full Text
- View/download PDF
25. Plasmodium falciparum erythrocyte invasion: combining function with immune evasion.
- Author
-
Gavin J Wright and Julian C Rayner
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
All the symptoms and pathology of malaria are caused by the intraerythrocytic stages of the Plasmodium parasite life cycle. Because Plasmodium parasites cannot replicate outside a host cell, their ability to recognize and invade erythrocytes is an essential step for both parasite survival and malaria pathogenesis. This makes invasion a conceptually attractive vaccine target, especially because it is one of the few stages when the parasite is directly exposed to the host humoral immune system. This apparent vulnerability, however, has been countered by the parasite, which has evolved sophisticated molecular mechanisms to evade the host immune response so that parasites asymptomatically replicate within immune individuals. These mechanisms include the expansion of parasite invasion ligands, resulting in multiple and apparently redundant invasion "pathways", highly polymorphic parasite surface proteins that are immunologically distinct, and parasite proteins which are poorly immunogenic. These formidable defences have so far thwarted attempts to develop an effective blood-stage vaccine, leading many to question whether there really is an exploitable chink in the parasite's immune evasion defences. Here, we review recent advances in the molecular understanding of the P. falciparum erythrocyte invasion field, discuss some of the challenges that have so far prevented the development of blood-stage vaccines, and conclude that the parasite invasion ligand RH5 represents an essential pinch point that might be vulnerable to vaccination.
- Published
- 2014
- Full Text
- View/download PDF
26. Semaphorin-7A is an erythrocyte receptor for P. falciparum merozoite-specific TRAP homolog, MTRAP.
- Author
-
S Josefin Bartholdson, Leyla Y Bustamante, Cecile Crosnier, Steven Johnson, Susan Lea, Julian C Rayner, and Gavin J Wright
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
The motility and invasion of Plasmodium parasites is believed to require a cytoplasmic actin-myosin motor associated with a cell surface ligand belonging to the TRAP (thrombospondin-related anonymous protein) family. Current models of invasion usually invoke the existence of specific receptors for the TRAP-family ligands on the surface of the host cell; however, the identities of these receptors remain largely unknown. Here, we identify the GPI-linked protein Semaphorin-7A (CD108) as an erythrocyte receptor for the P. falciparum merozoite-specific TRAP homolog (MTRAP) by using a systematic screening approach designed to detect extracellular protein interactions. The specificity of the interaction was demonstrated by showing that binding was saturable and by quantifying the equilibrium and kinetic biophysical binding parameters using surface plasmon resonance. We found that two MTRAP monomers interact via their tandem TSR domains with the Sema domains of a Semaphorin-7A homodimer. Known naturally-occurring polymorphisms in Semaphorin-7A did not quantitatively affect MTRAP binding nor did the presence of glycans on the receptor. Attempts to block the interaction during in vitro erythrocyte invasion assays using recombinant proteins and antibodies showed no significant inhibitory effect, suggesting the inaccessibility of the complex to proteinaceous blocking agents. These findings now provide important experimental evidence to support the model that parasite TRAP-family ligands interact with specific host receptors during cellular invasion.
- Published
- 2012
- Full Text
- View/download PDF
27. The alveolin IMC1h is required for normal ookinete and sporozoite motility behaviour and host colonisation in Plasmodium berghei.
- Author
-
Katrin Volkmann, Claudia Pfander, Charlotte Burstroem, Malika Ahras, David Goulding, Julian C Rayner, Friedrich Frischknecht, Oliver Billker, and Mathieu Brochet
- Subjects
Medicine ,Science - Abstract
Alveolins, or inner membrane complex (IMC) proteins, are components of the subpellicular network that forms a structural part of the pellicle of malaria parasites. In Plasmodium berghei, deletions of three alveolins, IMC1a, b, and h, each resulted in reduced mechanical strength and gliding velocity of ookinetes or sporozoites. Using time lapse imaging, we show here that deletion of IMC1h (PBANKA_143660) also has an impact on the directionality and motility behaviour of both ookinetes and sporozoites. Despite their marked motility defects, sporozoites lacking IMC1h were able to invade mosquito salivary glands, allowing us to investigate the role of IMC1h in colonisation of the mammalian host. We show that IMC1h is essential for sporozoites to progress through the dermis in vivo but does not play a significant role in hepatoma cell transmigration and invasion in vitro. Colocalisation of IMC1h with the residual IMC in liver stages was detected up to 30 hours after infection and parasites lacking IMC1h showed developmental defects in vitro and a delayed onset of blood stage infection in vivo. Together, these results suggest that IMC1h is involved in maintaining the cellular architecture which supports normal motility behaviour, access of the sporozoites to the blood stream, and further colonisation of the mammalian host.
- Published
- 2012
- Full Text
- View/download PDF
28. Biochemical and functional analysis of two Plasmodium falciparum blood-stage 6-cys proteins: P12 and P41.
- Author
-
Tana Taechalertpaisarn, Cecile Crosnier, S Josefin Bartholdson, Anthony N Hodder, Jenny Thompson, Leyla Y Bustamante, Danny W Wilson, Paul R Sanders, Gavin J Wright, Julian C Rayner, Alan F Cowman, Paul R Gilson, and Brendan S Crabb
- Subjects
Medicine ,Science - Abstract
The genomes of Plasmodium parasites that cause malaria in humans, other primates, birds, and rodents all encode multiple 6-cys proteins. Distinct 6-cys protein family members reside on the surface at each extracellular life cycle stage and those on the surface of liver infective and sexual stages have been shown to play important roles in hepatocyte growth and fertilization respectively. However, 6-cys proteins associated with the blood-stage forms of the parasite have no known function. Here we investigate the biochemical nature and function of two blood-stage 6-cys proteins in Plasmodium falciparum, the most pathogenic species to afflict humans. We show that native P12 and P41 form a stable heterodimer on the infective merozoite surface and are secreted following invasion, but could find no evidence that this complex mediates erythrocyte-receptor binding. That P12 and P41 do not appear to have a major role as adhesins to erythrocyte receptors was supported by the observation that antisera to these proteins did not substantially inhibit erythrocyte invasion. To investigate other functional roles for these proteins their genes were successfully disrupted in P. falciparum, however P12 and P41 knockout parasites grew at normal rates in vitro and displayed no other obvious phenotypic changes. It now appears likely that these blood-stage 6-cys proteins operate as a pair and play redundant roles either in erythrocyte invasion or in host-immune interactions.
- Published
- 2012
- Full Text
- View/download PDF
29. Phenotypic Screens Identify Genetic Factors Associated with Gametocyte Development in the Human Malaria Parasite Plasmodium falciparum
- Author
-
Jyotsna Chawla, Ilana Goldowitz, Jenna Oberstaller, Min Zhang, Camilla Valente Pires, Francesca Navarro, Lauriane Sollelis, Chengqi C. Q. Wang, Andreas Seyfang, Jeffrey Dvorin, Thomas D. Otto, Julian C. Rayner, Matthias Marti, and John H. Adams
- Subjects
Microbiology (medical) ,Infectious Diseases ,General Immunology and Microbiology ,Ecology ,Physiology ,Genetics ,Cell Biology - Abstract
Blocking human-to-vector transmission is an essential step toward malaria elimination. Gametocytes are solely responsible for achieving this transmission and represent an opportunity for therapeutic intervention.
- Published
- 2023
30. Chemogenomic Profiling of a Plasmodium falciparum Transposon Mutant Library Reveals Shared Effects of Dihydroartemisinin and Bortezomib on Lipid Metabolism and Exported Proteins
- Author
-
Camilla Valente Pires, Jenna Oberstaller, Chengqi Wang, Debora Casandra, Min Zhang, Jyotsna Chawla, Swamy Rakesh Adapa, Thomas D. Otto, Michael T. Ferdig, Julian C. Rayner, Rays H. Y. Jiang, and John H. Adams
- Subjects
Microbiology (medical) ,Infectious Diseases ,General Immunology and Microbiology ,Ecology ,Physiology ,Genetics ,Cell Biology - Abstract
Malaria control is seriously threatened by the emergence and spread of Plasmodium falciparum resistance to the leading antimalarial, artemisinin. The potent killing activity of artemisinin results from oxidative damage unleashed by free heme activation released by hemoglobin digestion.
- Published
- 2023
31. Lineage-informative microhaplotypes for spatio-temporal surveillance of Plasmodium vivax malaria parasites
- Author
-
Sasha V. Siegel, Roberto Amato, Hidayat Trimarsanto, Edwin Sutanto, Mariana Kleinecke, Kathryn Murie, Georgia Whitton, Aimee R. Taylor, James A. Watson, Mallika Imwong, Ashenafi Assefa, Awab Ghulam Rahim, Nguyen Hoang Chau, Tran Tinh Hien, Justin A Green, Gavin Koh, Nicholas J. White, Nicholas Day, Dominic P. Kwiatkowski, Julian C. Rayner, Ric N. Price, and Sarah Auburn
- Subjects
Article - Abstract
Challenges in understanding the origin of recurrentPlasmodium vivaxinfections constrains the surveillance of antimalarial efficacy and transmission of this neglected parasite. Recurrent infections within an individual may arise from activation of dormant liver stages (relapse), blood-stage treatment failure (recrudescence) or new inoculations (reinfection). Molecular inference of familial relatedness (identity-by-descent or IBD) based on whole genome sequence data, together with analysis of the intervals between parasitaemic episodes (“time-to-event” analysis), can help resolve the probable origin of recurrences. Whole genome sequencing of predominantly low-densityP. vivaxinfections is challenging, so an accurate and scalable genotyping method to determine the origins of recurrent parasitaemia would be of significant benefit. We have developed aP. vivaxgenome-wide informatics pipeline to select specific microhaplotype panels that can capture IBD within small, amplifiable segments of the genome. Using a global set of 615P. vivaxgenomes, we derived a panel of 100 microhaplotypes, each comprising 3-10 high frequency SNPs within HE= 0.70-0.81) and it captured 89% (273/307) of the polyclonal infections detected with genome-wide datasets. Using data simulations, we demonstrate lower error in estimating pairwise IBD using microhaplotypes, relative to traditional biallelic SNP barcodes. Our panel exhibited high accuracy in predicting the country of origin (median Matthew’s correlation coefficient >0.9 in 90% countries tested) and it also captured local infection outbreak and bottlenecking events. The informatics pipeline is available open-source and yields microhaplotypes that can be readily transferred to high-throughput amplicon sequencing assays for surveillance in malaria-endemic regions.
- Published
- 2023
32. Design and implementation of multiplexed amplicon sequencing panels to serve genomic epidemiology of infectious disease: A malaria case study
- Author
-
Emily LaVerriere, Philipp Schwabl, Manuela Carrasquilla, Aimee R. Taylor, Zachary M. Johnson, Meg Shieh, Ruchit Panchal, Timothy J. Straub, Rebecca Kuzma, Sean Watson, Caroline O. Buckee, Carolina M. Andrade, Silvia Portugal, Peter D. Crompton, Boubacar Traore, Julian C. Rayner, Vladimir Corredor, Kashana James, Horace Cox, Angela M. Early, Bronwyn L. MacInnis, Daniel E. Neafsey, LaVerriere, Emily [0000-0002-4750-4199], Schwabl, Philipp [0000-0002-1244-945X], Carrasquilla, Manuela [0000-0002-7953-4376], Shieh, Meg [0000-0002-0483-5000], Panchal, Ruchit [0000-0002-4664-4501], and Apollo - University of Cambridge Repository
- Subjects
relatedness ,amplicon sequencing ,Plasmodium falciparum ,malaria ,Genomics ,Communicable Diseases ,Article ,genotyping ,Malaria, Vivax ,Genetics ,Humans ,epidemiology ,Plasmodium vivax ,genome ,Ecology, Evolution, Behavior and Systematics ,Biotechnology - Abstract
Funder: British Council; Id: http://dx.doi.org/10.13039/501100000308, Funder: Broad Institute; Id: http://dx.doi.org/10.13039/100013114, Funder: National Institute of Allergy and Infectious Diseases; Id: http://dx.doi.org/10.13039/100000060, Funder: Bill and Melinda Gates Foundation; Id: http://dx.doi.org/10.13039/100000865, Funder: Division of Intramural Research, National Institute of Allergy and Infectious Diseases; Id: http://dx.doi.org/10.13039/100006492, Multiplexed PCR amplicon sequencing (AmpSeq) is an increasingly popular application for cost-effective monitoring of threatened species and managed wildlife populations, and shows strong potential for the genomic epidemiology of infectious disease. AmpSeq data from infectious microbes can inform disease control in multiple ways, such as by measuring drug resistance marker prevalence, distinguishing imported from local cases, and determining the effectiveness of therapeutics. We describe the design and comparative evaluation of two new AmpSeq assays for Plasmodium falciparum malaria parasites: a four-locus panel ("4CAST") composed of highly diverse antigens, and a 129-locus panel ("AMPLseq") composed of drug resistance markers, highly diverse loci for inferring relatedness, and a locus to detect Plasmodium vivax co-infection. We explore the performance of each panel in various public health use cases with in silico simulations as well as empirical experiments. The 4CAST panel appears highly suitable for evaluating the number of distinct parasite strains within samples (complexity of infection), showing strong performance across a wide range of parasitaemia levels without a DNA pre-amplification step. For relatedness inference, the larger AMPLseq panel performs similarly to two existing panels of comparable size, despite differences in the data and approach used for designing each panel. Finally, we describe an R package (paneljudge) that facilitates the design and comparative evaluation of genetic panels for relatedness estimation, and we provide general guidance on the design and implementation of AmpSeq panels for the genomic epidemiology of infectious disease.
- Published
- 2022
33. A molecular barcode and web-based data analysis tool to identify imported Plasmodium vivax malaria
- Author
-
Hidayat Trimarsanto, Roberto Amato, Richard D. Pearson, Edwin Sutanto, Rintis Noviyanti, Leily Trianty, Jutta Marfurt, Zuleima Pava, Diego F. Echeverry, Tatiana M. Lopera-Mesa, Lidia M. Montenegro, Alberto Tobón-Castaño, Matthew J. Grigg, Bridget Barber, Timothy William, Nicholas M. Anstey, Sisay Getachew, Beyene Petros, Abraham Aseffa, Ashenafi Assefa, Awab G. Rahim, Nguyen H. Chau, Tran T. Hien, Mohammad S. Alam, Wasif A. Khan, Benedikt Ley, Kamala Thriemer, Sonam Wangchuck, Yaghoob Hamedi, Ishag Adam, Yaobao Liu, Qi Gao, Kanlaya Sriprawat, Marcelo U. Ferreira, Moses Laman, Alyssa Barry, Ivo Mueller, Marcus V. G. Lacerda, Alejandro Llanos-Cuentas, Srivicha Krudsood, Chanthap Lon, Rezika Mohammed, Daniel Yilma, Dhelio B. Pereira, Fe E. J. Espino, Cindy S. Chu, Iván D. Vélez, Chayadol Namaik-larp, Maria F. Villegas, Justin A. Green, Gavin Koh, Julian C. Rayner, Eleanor Drury, Sónia Gonçalves, Victoria Simpson, Olivo Miotto, Alistair Miles, Nicholas J. White, Francois Nosten, Dominic P. Kwiatkowski, Ric N. Price, Sarah Auburn, Pearson, Richard D [0000-0002-7386-3566], Sutanto, Edwin [0000-0002-2997-4648], Marfurt, Jutta [0000-0002-3846-0825], Echeverry, Diego F [0000-0003-0301-4478], Tobón-Castaño, Alberto [0000-0002-1671-2649], Grigg, Matthew J [0000-0001-9914-8352], Alam, Mohammad S [0000-0001-8330-5499], Khan, Wasif A [0000-0002-7650-8068], Ley, Benedikt [0000-0002-5734-0845], Mueller, Ivo [0000-0001-6554-6889], Lacerda, Marcus VG [0000-0003-3374-9985], Pereira, Dhelio B [0000-0002-7761-5498], Espino, Fe EJ [0000-0003-1690-1711], Koh, Gavin [0000-0002-7336-1566], Rayner, Julian C [0000-0002-9835-1014], Drury, Eleanor [0000-0002-9887-6961], Miotto, Olivo [0000-0001-8060-6771], Nosten, Francois [0000-0002-7951-0745], Kwiatkowski, Dominic P [0000-0002-5023-0176], Price, Ric N [0000-0003-2000-2874], Auburn, Sarah [0000-0002-4638-536X], and Apollo - University of Cambridge Repository
- Subjects
Likelihood Functions ,Internet ,Molecular medicine ,45 ,Epidemiology ,692/308/2056 ,article ,Medicine (miscellaneous) ,631/114/1305 ,General Biochemistry, Genetics and Molecular Biology ,692/4017 ,Malaria ,Genetics research ,Machine learning ,Malaria, Vivax ,692/699/255/1629 ,Humans ,PLASMODIUM ,692/308/174 ,Plasmodium vivax ,General Agricultural and Biological Sciences - Abstract
Funder: HT was supported by a Charles Darwin University International PhD Scholarship (CDIPS), Funder: Malaysian Ministry of Health (BP00500420), Traditionally, patient travel history has been used to distinguish imported from autochthonous malaria cases, but the dormant liver stages of Plasmodium vivax confound this approach. Molecular tools offer an alternative method to identify, and map imported cases. Using machine learning approaches incorporating hierarchical fixation index and decision tree analyses applied to 799 P. vivax genomes from 21 countries, we identified 33-SNP, 50-SNP and 55-SNP barcodes (GEO33, GEO50 and GEO55), with high capacity to predict the infection's country of origin. The Matthews correlation coefficient (MCC) for an existing, commonly applied 38-SNP barcode (BR38) exceeded 0.80 in 62% countries. The GEO panels outperformed BR38, with median MCCs > 0.80 in 90% countries at GEO33, and 95% at GEO50 and GEO55. An online, open-access, likelihood-based classifier framework was established to support data analysis (vivaxGEN-geo). The SNP selection and classifier methods can be readily amended for other use cases to support malaria control programs.
- Published
- 2022
34. Barcoding Genetically Distinct Plasmodium falciparum Strains for Comparative Assessment of Fitness and Antimalarial Drug Resistance
- Author
-
Manuela Carrasquilla, Ndey F. Drammeh, Mukul Rawat, Theo Sanderson, Zenon Zenonos, Julian C. Rayner, and Marcus C. S. Lee
- Subjects
Antimalarials ,Virology ,Plasmodium falciparum ,Drug Resistance ,Protozoan Proteins ,Genetic Fitness ,Complex Mixtures ,Microbiology ,Artemisinins - Abstract
The repeated emergence of antimalarial drug resistance in Plasmodium falciparum, including to the current frontline antimalarial artemisinin, is a perennial problem for malaria control. Next-generation sequencing has greatly accelerated the identification of polymorphisms in resistance-associated genes but has also highlighted the need for more sensitive and accurate laboratory tools to profile current and future antimalarials and to quantify the impact of drug resistance acquisition on parasite fitness. The interplay of fitness and drug response is of fundamental importance in understanding why particular genetic backgrounds are better at driving the evolution of drug resistance in natural populations, but the impact of parasite fitness landscapes on the epidemiology of drug resistance has typically been laborious to accurately quantify in the lab, with assays being limited in accuracy and throughput. Here we present a scalable method to profile fitness and drug response of genetically distinct P. falciparum strains with well-described sensitivities to several antimalarials. We leverage CRISPR/Cas9 genome-editing and barcode sequencing to track unique barcodes integrated into a nonessential gene (
- Published
- 2022
35. The structure of a Plasmodium vivax Tryptophan Rich Antigen suggests a lipid binding function for a pan-Plasmodium multi-gene family
- Author
-
Prasun Kundu, Deboki Naskar, Shannon McKie, Sheena Dass, Usheer Kanjee, Viola Introini, Marcelo U. Ferreira, Manoj Duraisingh, Janet Deane, and Julian C. Rayner
- Abstract
Tryptophan Rich Antigens (TRAgs) are encoded by a multi-gene family in all Plasmodium species, significantly expanded in P. vivax, but their function is not currently known. We show that multiple P. vivax TRAgs are expressed on the merozoite surface and that one, PVP01_0000100 binds red blood cells with a strong preference for reticulocytes. Solving the structure of the C-terminal tryptophan rich domain that defines the TRAg family revealed a three-helical bundle that is conserved across Plasmodium and has homology with lipid-binding BAR domains involved in membrane remodelling. Biochemical assays confirmed that this domain has lipid binding activity with preference for sulfatide, a glycosphingolipid present in the outer leaflet of plasma membranes. Deletion of the putative orthologue in P. knowlesi, PKNH_1300500, impacts invasion in reticulocytes, suggesting a role for membrane remodelling during this essential process. Together, this work suggests a molecular function for the TRAg family for the first time.
- Published
- 2022
36. The erythrocyte membrane properties of beta thalassaemia heterozygotes and their consequences for Plasmodium falciparum invasion
- Author
-
Viola Introini, Alejandro Marin-Menendez, Guilherme Nettesheim, Yen-Chun Lin, Silvia N. Kariuki, Adrian L Smith, Letitia Jean, John N. Brewin, David C. Rees, Pietro Cicuta, Julian C. Rayner, Bridget S. Penman, Introini, Viola [0000-0001-9012-4696], Marin-Menendez, Alejandro [0000-0002-4903-6392], Nettesheim, Guilherme [0000-0001-7464-4333], Kariuki, Silvia N [0000-0003-0801-5285], Cicuta, Pietro [0000-0002-9193-8496], Rayner, Julian C [0000-0002-9835-1014], Penman, Bridget S [0000-0001-9803-5209], Apollo - University of Cambridge Repository, Rayner, Julian [0000-0002-9835-1014], Kariuki, Silvia [0000-0003-0801-5285], Jean, Letitia [0000-0002-6450-7148], and Rees, David [0000-0003-3647-1050]
- Subjects
2 Aetiology ,Heterozygote ,Multidisciplinary ,631/326/421 ,Erythrocyte Membrane ,Plasmodium falciparum ,beta-Thalassemia ,article ,FOS: Health sciences ,631/250/255/1629 ,QR ,Malaria ,Vector-Borne Diseases ,Rare Diseases ,Infectious Diseases ,parasitic diseases ,2.1 Biological and endogenous factors ,Humans ,Malaria, Falciparum ,Infection ,RC - Abstract
Funder: EPSRC, Funder: Sackler fellowship, Malaria parasites such as Plasmodium falciparum have exerted formidable selective pressures on the human genome. Of the human genetic variants associated with malaria protection, beta thalassaemia (a haemoglobinopathy) was the earliest to be associated with malaria prevalence. However, the malaria protective properties of beta thalassaemic erythrocytes remain unclear. Here we studied the mechanics and surface protein expression of beta thalassaemia heterozygous erythrocytes, measured their susceptibility to P. falciparum invasion, and calculated the energy required for merozoites to invade them. We found invasion-relevant differences in beta thalassaemic cells versus matched controls, specifically: elevated membrane tension, reduced bending modulus, and higher levels of expression of the major invasion receptor basigin. However, these differences acted in opposition to each other with respect to their likely impact on invasion, and overall we did not observe beta thalassaemic cells to have lower P. falciparum invasion efficiency for any of the strains tested.
- Published
- 2022
37. An open dataset of
- Author
-
Ishag, Adam, Mohammad Shafiul, Alam, Sisay, Alemu, Chanaki, Amaratunga, Roberto, Amato, Voahangy, Andrianaranjaka, Nicholas M, Anstey, Abraham, Aseffa, Elizabeth, Ashley, Ashenafi, Assefa, Sarah, Auburn, Bridget E, Barber, Alyssa, Barry, Dhelio, Batista Pereira, Jun, Cao, Nguyen Hoang, Chau, Kesinee, Chotivanich, Cindy, Chu, Arjen M, Dondorp, Eleanor, Drury, Diego F, Echeverry, Berhanu, Erko, Fe, Espino, Rick, Fairhurst, Abdul, Faiz, María, Fernanda Villegas, Qi, Gao, Lemu, Golassa, Sonia, Goncalves, Matthew J, Grigg, Yaghoob, Hamedi, Tran Tinh, Hien, Ye, Htut, Kimberly J, Johnson, Nadira, Karunaweera, Wasif, Khan, Srivicha, Krudsood, Dominic P, Kwiatkowski, Marcus, Lacerda, Benedikt, Ley, Pharath, Lim, Yaobao, Liu, Alejandro, Llanos-Cuentas, Chanthap, Lon, Tatiana, Lopera-Mesa, Jutta, Marfurt, Pascal, Michon, Olivo, Miotto, Rezika, Mohammed, Ivo, Mueller, Chayadol, Namaik-Larp, Paul N, Newton, Thuy-Nhien, Nguyen, Francois, Nosten, Rintis, Noviyanti, Zuleima, Pava, Richard D, Pearson, Beyene, Petros, Aung P, Phyo, Ric N, Price, Sasithon, Pukrittayakamee, Awab Ghulam, Rahim, Milijaona, Randrianarivelojosia, Julian C, Rayner, Angela, Rumaseb, Sasha V, Siegel, Victoria J, Simpson, Kamala, Thriemer, Alberto, Tobon-Castano, Hidayat, Trimarsanto, Marcelo, Urbano Ferreira, Ivan D, Vélez, Sonam, Wangchuk, Thomas E, Wellems, Nicholas J, White, Timothy, William, Maria F, Yasnot, and Daniel, Yilma
- Abstract
This report describes the MalariaGEN Pv4 dataset, a new release of curated genome variation data on 1,895 samples of
- Published
- 2022
38. Barcoding genetically distinctPlasmodium falciparumstrains for comparative assessment of fitness and antimalarial drug resistance
- Author
-
Manuela Carrasquilla, Ndey Fatou Drammeh, Mukul Rawat, Theo Sanderson, Zenon Zenonos, Julian C Rayner, and Marcus CS Lee
- Abstract
The repeated emergence of antimalarial drug resistance inPlasmodium falciparum, including to the current frontline antimalarial artemisinin, is a perennial problem for malaria control. Nextgeneration sequencing has greatly accelerated the identification of polymorphisms in resistance-associated genes, but has also highlighted the need for more sensitive and accurate laboratory tools to profile current and future antimalarials, and to quantify the impact of drug resistance acquisition on parasite fitness. The interplay of fitness and drug response is of fundamental importance in understanding why particular genetic backgrounds are better at driving the evolution of drug resistance in natural populations, but the impact of parasite fitness landscapes on the epidemiology of drug resistance has typically been laborious to accurately quantify in the lab, with assays being limited in accuracy and throughput. Here we present a scalable method to profile fitness and drug response of genetically distinctP. falciparumstrains with well-described sensitivities to several antimalarials. We leverage CRISPR/Cas9 genome-editing and barcode sequencing to track unique barcodes integrated into a non-essential gene (pfrh3). We validate this approach in multiplex competitive growth assays of three strains with distinct geographical origins. Furthermore, we demonstrate that this method can be a powerful approach for tracking artemisinin response as it can identify an artemisinin resistant strain within a mix of multiple parasite lines, suggesting an approach for scaling the laborious ring-stage survival assay (RSA) across libraries of barcoded parasite lines. Overall, we present a novel high-throughput method for multiplexed competitive growth assays to evaluate parasite fitness and drug response
- Published
- 2022
39. Biophysical Tools and Concepts Enable Understanding of Asexual Blood Stage Malaria
- Author
-
Viola Introini, Matt A. Govendir, Julian C. Rayner, Pietro Cicuta, Maria Bernabeu, Rayner, Julian [0000-0002-9835-1014], and Apollo - University of Cambridge Repository
- Subjects
Microbiology (medical) ,cytoadhesion ,Plasmodium ,Life Cycle Stages ,Erythrocytes ,Immunology ,Plasmodium falciparum ,microfluidics ,Protozoan Proteins ,imaging ,mechanobiology ,Microbiology ,Malaria ,Infectious Diseases ,biophysics ,parasitic diseases ,Humans - Abstract
Forces and mechanical properties of cells and tissues set constraints on biological functions, and are key determinants of human physiology. Changes in cell mechanics may arise from disease, or directly contribute to pathogenesis. Malaria gives many striking examples. Plasmodium parasites, the causative agents of malaria, are single-celled organisms that cannot survive outside their hosts; thus, thost-pathogen interactions are fundamental for parasite’s biological success and to the host response to infection. These interactions are often combinations of biochemical and mechanical factors, but most research focuses on the molecular side. However, Plasmodium infection of human red blood cells leads to changes in their mechanical properties, which has a crucial impact on disease pathogenesis because of the interaction of infected red blood cells with other human tissues through various adhesion mechanisms, which can be probed and modelled with biophysical techniques. Recently, natural polymorphisms affecting red blood cell biomechanics have also been shown to protect human populations, highlighting the potential of understanding biomechanical factors to inform future vaccines and drug development. Here we review biophysical techniques that have revealed new aspects of Plasmodium falciparum invasion of red blood cells and cytoadhesion of infected cells to the host vasculature. These mechanisms occur differently across Plasmodium species and are linked to malaria pathogenesis. We highlight promising techniques from the fields of bioengineering, immunomechanics, and soft matter physics that could be beneficial for studying malaria. Some approaches might also be applied to other phases of the malaria lifecycle and to apicomplexan infections with complex host-pathogen interactions.
- Published
- 2022
40. Refining the transcriptome of the human malaria parasite Plasmodium falciparum using amplification-free RNA-seq
- Author
-
Timothy J. Russell, Lia Chappell, Lindsey M. Orchard, Julian C. Rayner, Philipp Ross, Thomas D. Otto, Manuel Llinás, Matthew Berriman, Llinás, Manuel [0000-0002-6173-5882], and Apollo - University of Cambridge Repository
- Subjects
Untranslated region ,lcsh:QH426-470 ,lcsh:Biotechnology ,Plasmodium falciparum ,Protozoan Proteins ,RNA-Seq ,Computational biology ,Genome ,Eukaryote microbial genomics ,03 medical and health sciences ,0302 clinical medicine ,Species Specificity ,Transcription (biology) ,Complementary DNA ,lcsh:TP248.13-248.65 ,Genetics ,Humans ,RNA, Messenger ,Malaria, Falciparum ,3' Untranslated Regions ,030304 developmental biology ,Life Cycle Stages ,0303 health sciences ,biology ,Gene Expression Profiling ,Promoter ,biology.organism_classification ,3. Good health ,lcsh:Genetics ,DNA microarray ,5' Untranslated Regions ,Nucleic Acid Amplification Techniques ,030217 neurology & neurosurgery ,Biotechnology ,Research Article - Abstract
Background Plasmodium parasites undergo several major developmental transitions during their complex lifecycle, which are enabled by precisely ordered gene expression programs. Transcriptomes from the 48-h blood stages of the major human malaria parasite Plasmodium falciparum have been described using cDNA microarrays and RNA-seq, but these assays have not always performed well within non-coding regions, where the AT-content is often 90–95%. Results We developed a directional, amplification-free RNA-seq protocol (DAFT-seq) to reduce bias against AT-rich cDNA, which we have applied to three strains of P. falciparum (3D7, HB3 and IT). While strain-specific differences were detected, overall there is strong conservation between the transcriptional profiles. For the 3D7 reference strain, transcription was detected from 89% of the genome, with over 78% of the genome transcribed into mRNAs. We also find that transcription from bidirectional promoters frequently results in non-coding, antisense transcripts. These datasets allowed us to refine the 5′ and 3′ untranslated regions (UTRs), which can be variable, long (> 1000 nt), and often overlap those of adjacent transcripts. Conclusions The approaches applied in this study allow a refined description of the transcriptional landscape of P. falciparum and demonstrate that very little of the densely packed P. falciparum genome is inactive or redundant. By capturing the 5′ and 3′ ends of mRNAs, we reveal both constant and dynamic use of transcriptional start sites across the intraerythrocytic developmental cycle that will be useful in guiding the definition of regulatory regions for use in future experimental gene expression studies.
- Published
- 2020
41. Probing SARS-CoV-2-positive plasma to identify potential factors correlating with mild COVID-19 in Ghana, West Africa
- Author
-
Kesego Tapela, Fatima O. Oyawoye, Charles Ochieng’ Olwal, Precious C. Opurum, Jones Amo Amponsah, Kekeli Aku Lumor Segbedzi, Becky Tetteh, Frederick Kumi-Ansah, Joe K. Mutungi, Evangeline Obodai, Emmanuella Amoako, Seth Agyemang, Nicaise Tuikue Ndam, William Kwabena Ampofo, Julian C. Rayner, Gordon A. Awandare, Lily Paemka, Yaw Bediako, and Peter Kojo Quashie
- Subjects
Interleukin 1 Receptor Antagonist Protein ,Interleukin-6 ,SARS-CoV-2 ,Interleukin-8 ,COVID-19 ,Cytokines ,Humans ,General Medicine ,Ghana ,Biomarkers ,ABO Blood-Group System ,Uncategorized - Abstract
Background West Africa has recorded a relatively higher proportion of asymptomatic coronavirus disease 2019 (COVID-19) cases than the rest of the world, and West Africa-specific host factors could play a role in this discrepancy. Here, we assessed the association between COVID-19 severity among Ghanaians with their immune profiles and ABO blood groups. Methods Plasma samples were obtained from Ghanaians PCR-confirmed severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-positive individuals. The participants were categorized into symptomatic and asymptomatic cases. Cytokine profiling and antibody quantification were performed using Luminex™ multiplex assay whereas antigen-driven agglutination assay was used to assess the ABO blood groups. Immune profile levels between symptomatic and asymptomatic groups were compared using the two-tailed Mann-Whitney U test. Multiple comparisons of cytokine levels among and between days were tested using Kruskal-Wallis with Dunn’s post hoc test. Correlations within ABO blood grouping (O’s and non-O’s) and between cytokines were determined using Spearman correlations. Logistic regression analysis was performed to assess the association of various cytokines with asymptomatic phenotype. Results There was a trend linking blood group O to reduced disease severity, but this association was not statistically significant. Generally, symptomatic patients displayed significantly (p p Conclusions The findings suggest that genetic background and pre-existing immune response patterns may in part shape the nature of the symptomatic response against COVID-19 in a West African population. This study offers clear directions to be explored further in larger studies.
- Published
- 2022
- Full Text
- View/download PDF
42. Controlled human malaria infection with a clone of Plasmodium vivax with high-quality genome assembly
- Author
-
Geneviève M. Labbé, Eerik Aunin, Yrene Themistocleous, Jee Sun Cho, Raquel Lopez Ramon, Julian C. Rayner, Baktash Khozoee, Chayanut Suansomjit, Sarah E. Silk, Doris Quinkert, Alison Kemp, Thomas D. Otto, Iona J. Taylor, Alison M. Lawrie, Simon J. Draper, Angela M. Minassian, Thomas A. Rawlinson, Marija Zaric, Celia Mitton, Jason C. Sousa, Jetsumon Sattabongkot, Andrew M. Blagborough, Sumi Biswas, Kimberly J. Johnson, Florian Bach, Chalermpon Kumpitak, Adam J. Reid, Margaux Mulatier, Nongnuj Maneechai, Ian D. Poulton, Katherine J. Ellis, Nattawan Rachaphaew, Nick J. Edwards, Fernando Ramos Lopez, Philip J Spence, Carolyn M. Nielsen, Wanlapa Roobsoong, Arianna Marini, Kirsty McHugh, Megan Baker, David J. Roberts, Mimi M. Hou, Tianrat Piteekan, Jordan R. Barrett, Fay L. Nugent, Marini, Arianna [0000-0003-3416-7362], Rayner, Julian [0000-0002-9835-1014], and Apollo - University of Cambridge Repository
- Subjects
Male ,Infectious disease ,Genome ,biology ,Plasmodium vivax ,Clone (cell biology) ,Plasmodium falciparum ,General Medicine ,Parasitemia ,medicine.disease ,biology.organism_classification ,Virology ,Healthy Volunteers ,Malaria ,Antigen ,Infectious disease (medical specialty) ,Resource and Technical Advance ,parasitic diseases ,medicine ,Animals ,Humans ,Malaria, Falciparum - Abstract
Controlled human malaria infection (CHMI) provides a highly informative means to investigate host-pathogen interactions and enable in vivo proof-of-concept efficacy testing of new drugs and vaccines. However, unlike Plasmodium falciparum, well-characterized P. vivax parasites that are safe and suitable for use in modern CHMI models are limited. Here, two healthy malaria-naïve UK adults with universal donor blood group were safely infected with a clone of P. vivax from Thailand by mosquito-bite CHMI. Parasitemia developed in both volunteers and, prior to treatment, each volunteer donated blood to produce a cryopreserved stabilate of infected red blood cells. Following stringent safety screening, the parasite stabilate from one of these donors ("PvW1") was thawed and used to inoculate six healthy malaria-naïve UK adults by blood-stage CHMI, at three different dilutions. Parasitemia developed in all volunteers, who were then successfully drug treated. PvW1 parasite DNA was isolated and sequenced to produce a high quality genome assembly by using a hybrid assembly method. We analysed leading vaccine candidate antigens and multigene families, including the Vivax interspersed repeat (VIR) genes of which we identified 1145 in the PvW1 genome. Our genomic analysis will guide future assessment of candidate vaccines and drugs, as well as experimental medicine studies.
- Published
- 2021
43. Design and implementation of multiplexed amplicon sequencing panels to serve genomic epidemiology of infectious disease: a malaria case study
- Author
-
Manuela Carrasquilla, Ruchit Panchal, Sean Watson, Caroline O. Buckee, Daniel E. Neafsey, Kashana James, Julian C. Rayner, Horace Cox, Emily LaVerriere, Bronwyn MacInnis, Peter D. Crompton, Angela M. Early, Carolina M. Andrade, Rebecca Kuzma, Aimee R. Taylor, Boubacar Traore, Silvia Portugal, Timothy J. Straub, Meg Shieh, Vladimir Corredor, Zachary M. Johnson, and Philipp Schwabl
- Subjects
biology ,Infectious disease (medical specialty) ,In silico ,Plasmodium vivax ,medicine ,Locus (genetics) ,Plasmodium falciparum ,Computational biology ,Drug resistance ,Parasitemia ,medicine.disease ,biology.organism_classification ,Malaria - Abstract
Multiplexed PCR amplicon sequencing (AmpSeq) is an increasingly popular application for cost-effective monitoring of threatened species and managed wildlife populations, and shows strong potential for genomic epidemiology of infectious disease. AmpSeq data for infectious microbes can inform disease control in multiple ways, including measuring drug resistance marker prevalence, distinguishing imported from local cases, and determining the effectiveness of therapeutics. We describe the design and comparative evaluation of two new AmpSeq assays for Plasmodium falciparum malaria parasites: a four-locus panel (‘4CAST’) composed of highly diverse antigens, and a 129-locus panel (‘AMPLseq’) composed of drug resistance markers, highly diverse loci for measuring relatedness, and a locus to detect Plasmodium vivax co-infections. We explore the performance of each panel in various public health use cases with in silico simulations as well as empirical experiments. We find that the smaller 4CAST panel performs reliably across a wide range of parasitemia levels without DNA pre-amplification, and could be highly informative for evaluating the number of distinct parasite strains within samples (complexity of infection), and distinguishing recrudescent infections from new infections in therapeutic efficacy studies. The AMPLseq panel performs similarly to two existing panels of comparable size for relatedness measurement, despite differences in the data and approach used for designing each panel. Finally, we describe an R package (paneljudge) that facilitates design and comparative evaluation of AmpSeq panels for relatedness estimation, and we provide general guidance on the design and implementation of AmpSeq panels for genomic epidemiology of infectious disease.
- Published
- 2021
44. The apicoplast link to fever-survival and artemisinin-resistance in the malaria parasite
- Author
-
Debora Casandra, Phaedra Thomas, John H. Adams, Michael T. Ferdig, Katrina A. Button-Simons, Shulin Xu, Sandhya Boyapalle, Swamy R. Adapa, Min Zhang, Rays H. Y. Jiang, Chengqi Wang, Julian C. Rayner, Thomas D. Otto, Matthew Mayho, Jenna Oberstaller, Zhang, Min [0000-0002-5443-0992], Oberstaller, Jenna [0000-0002-3033-2212], Thomas, Phaedra [0000-0003-3861-1213], Button-Simons, Katrina [0000-0001-5248-7939], Rayner, Julian C [0000-0002-9835-1014], Adams, John H [0000-0003-3707-7979], Apollo - University of Cambridge Repository, Adapa, Swamy R [0000-0003-3313-2782], Rayner, Julian C. [0000-0002-9835-1014], and Adams, John H. [0000-0003-3707-7979]
- Subjects
0301 basic medicine ,Transcription, Genetic ,Drug Resistance ,General Physics and Astronomy ,Plasmodium ,Genome ,0302 clinical medicine ,Parasite physiology ,Artemisinin ,Malaria, Falciparum ,Genetics ,631/326/417/2552 ,0303 health sciences ,631/326/417/2551 ,Multidisciplinary ,biology ,Temperature ,45/77 ,humanities ,Artemisinins ,3. Good health ,Phenotype ,38/39 ,medicine.drug ,Signal Transduction ,Fever ,Science ,Phenotypic screening ,Plasmodium falciparum ,13/106 ,45/23 ,Apicoplasts ,General Biochemistry, Genetics and Molecular Biology ,Article ,38/91 ,03 medical and health sciences ,parasitic diseases ,medicine ,692/699/255/1629 ,Animals ,Parasites ,631/326/417/2546 ,Gene ,030304 developmental biology ,Apicoplast ,030306 microbiology ,Terpenes ,Parasite genomics ,General Chemistry ,biology.organism_classification ,medicine.disease ,Malaria ,030104 developmental biology ,Gene Expression Regulation ,Mutation ,Unfolded Protein Response ,030217 neurology & neurosurgery ,Heat-Shock Response ,Parasite host response - Abstract
Funder: U.S. Department of Health & Human Services | NIH | National Institute of Allergy and Infectious Diseases (NIAID), The emergence and spread of Plasmodium falciparum parasites resistant to front-line antimalarial artemisinin-combination therapies (ACT) threatens to erase the considerable gains against the disease of the last decade. Here, we develop a large-scale phenotypic screening pipeline and use it to carry out a large-scale forward-genetic phenotype screen in P. falciparum to identify genes allowing parasites to survive febrile temperatures. Screening identifies more than 200 P. falciparum mutants with differential responses to increased temperature. These mutants are more likely to be sensitive to artemisinin derivatives as well as to heightened oxidative stress. Major processes critical for P. falciparum tolerance to febrile temperatures and artemisinin include highly essential, conserved pathways associated with protein-folding, heat shock and proteasome-mediated degradation, and unexpectedly, isoprenoid biosynthesis, which originated from the ancestral genome of the parasite’s algal endosymbiont-derived plastid, the apicoplast. Apicoplast-targeted genes in general are upregulated in response to heat shock, as are other Plasmodium genes with orthologs in plant and algal genomes. Plasmodium falciparum parasites appear to exploit their innate febrile-response mechanisms to mediate resistance to artemisinin. Both responses depend on endosymbiont-derived genes in the parasite’s genome, suggesting a link to the evolutionary origins of Plasmodium parasites in free-living ancestors.
- Published
- 2021
45. Controlled human malaria infection with PvW1 – a new clone of Plasmodium vivax with high quality genome assembly
- Author
-
Raquel Lopez Ramon, Chayanut Suansomjit, Fernando Ramos Lopez, Angela M. Minassian, Philip J Spence, Margaux Mulatier, Baktash Khozoee, Florian Bach, Carolyn M. Nielsen, Katherine J. Ellis, Chalermpon Kumpitak, Megan Baker, Jordan R. Barrett, Nattawan Rachaphaew, Sarah E. Silk, Kimberly J. Johnson, Ian D. Poulton, Doris Quinkert, Nick J. Edwards, Mimi M. Hou, Alison M. Lawrie, Yrene Themistocleous, Tianrat Piteekan, Simon J. Draper, Jee Sun Cho, Wanlapa Roobsoong, Adam J. Reid, Arianna Marini, Nongnuj Maneechai, Geneviève M. Labbé, Jetsumon Sattabongkot, Celia Mitton, Iona J. Taylor, Julian C. Rayner, Fay L. Nugent, Jason C. Sousa, Thomas D. Otto, David J. Roberts, Alison Kemp, Andrew M. Blagborough, Thomas A. Rawlinson, Marija Zaric, Sumi Biswas, and Eerik Aunin
- Subjects
Plasmodium vivax ,Clone (cell biology) ,Plasmodium falciparum ,Parasitemia ,Biology ,medicine.disease ,biology.organism_classification ,Genome ,Virology ,Antigen ,parasitic diseases ,medicine ,Gene ,Malaria - Abstract
Controlled human malaria infection (CHMI) provides a highly informative means to investigate host-pathogen interactions and enable in vivo proof-of-concept efficacy testing of new drugs and vaccines. However, unlike Plasmodium falciparum, well-characterized P. vivax parasites that are safe and suitable for use in modern CHMI models are limited. Here, two healthy malaria-naïve UK adults with universal donor blood group were safely infected with a clone of P. vivax from Thailand by mosquito-bite CHMI. Parasitemia developed in both volunteers and, prior to treatment, each volunteer donated blood to produce a cryopreserved stabilate of infected red blood cells. Following stringent safety screening, the parasite stabilate from one of these donors (“PvW1”) was thawed and used to inoculate six healthy malaria-naïve UK adults by blood-stage CHMI, at three different dilutions. Parasitemia developed in all volunteers, who were then successfully drug treated. PvW1 parasite DNA was isolated and sequenced to produce a high quality genome assembly by using a hybrid assembly method. We analysed leading vaccine candidate antigens and multigene families, including the Vivax interspersed repeat (VIR) genes of which we identified 1145 in the PvW1 genome. Our genomic analysis will guide future assessment of candidate vaccines and drugs, as well as experimental medicine studies.
- Published
- 2021
46. Using Plasmodium knowlesi as a model for screening Plasmodium vivax blood-stage malaria vaccine targets reveals new candidates
- Author
-
Rachael Coyle, Duncan N. Ndegwa, Julian C. Rayner, Alejandro Marin-Menendez, Jessica B. Hostetler, Lisa H. Verzier, Theo Sanderson, Sophie H. Adjalley, Kioko Mwikali, Prasun Kundu, Ndegwa, Duncan N. [0000-0002-6980-9102], Marin-Menendez, Alejandro [0000-0002-4903-6392], Sanderson, Theo [0000-0003-4177-2851], Mwikali, Kioko [0000-0003-4252-0744], Verzier, Lisa H. [0000-0002-5518-8157], Coyle, Rachael [0000-0001-5838-1086], Adjalley, Sophie [0000-0001-6658-1707], Rayner, Julian C. [0000-0002-9835-1014], Apollo - University of Cambridge Repository, Ndegwa, Duncan N [0000-0002-6980-9102], Verzier, Lisa H [0000-0002-5518-8157], and Rayner, Julian C [0000-0002-9835-1014]
- Subjects
Plasmodium ,Physiology ,Plasmodium vivax ,Protozoan Proteins ,Antibodies, Protozoan ,Biochemistry ,Medical Conditions ,0302 clinical medicine ,Animal Cells ,Immune Physiology ,Red Blood Cells ,Biology (General) ,Cells, Cultured ,Protozoans ,Vaccines ,0303 health sciences ,Immune System Proteins ,biology ,Malarial Parasites ,Eukaryota ,Genomics ,3. Good health ,Infectious Diseases ,Plasmodium knowlesi ,Cellular Types ,Antibody ,Research Article ,Infectious Disease Control ,QH301-705.5 ,030231 tropical medicine ,Immunology ,Antigens, Protozoan ,Microbiology ,Antibodies ,03 medical and health sciences ,Antigen ,Virology ,Parasite Groups ,Malaria Vaccines ,parasitic diseases ,Parasitic Diseases ,Malaria, Vivax ,Genetics ,medicine ,Humans ,Molecular Biology ,030304 developmental biology ,Medicine and health sciences ,Blood Cells ,Biology and life sciences ,Merozoites ,Reverse vaccinology ,Organisms ,Proteins ,Cell Biology ,RC581-607 ,medicine.disease ,biology.organism_classification ,Parasitic Protozoans ,Polyclonal antibodies ,biology.protein ,Parasitology ,Immunologic diseases. Allergy ,Apicomplexa ,Malaria - Abstract
Plasmodium vivax is responsible for the majority of malaria cases outside Africa. Unlike P. falciparum, the P. vivax life-cycle includes a dormant liver stage, the hypnozoite, which can cause infection in the absence of mosquito transmission. An effective vaccine against P. vivax blood stages would limit symptoms and pathology from such recurrent infections, and therefore could play a critical role in the control of this species. Vaccine development in P. vivax, however, lags considerably behind P. falciparum, which has many identified targets with several having transitioned to Phase II testing. By contrast only one P. vivax blood-stage vaccine candidate based on the Duffy Binding Protein (PvDBP), has reached Phase Ia, in large part because the lack of a continuous in vitro culture system for P. vivax limits systematic screening of new candidates. We used the close phylogenetic relationship between P. vivax and P. knowlesi, for which an in vitro culture system in human erythrocytes exists, to test the scalability of systematic reverse vaccinology to identify and prioritise P. vivax blood-stage targets. A panel of P. vivax proteins predicted to function in erythrocyte invasion were expressed as full-length recombinant ectodomains in a mammalian expression system. Eight of these antigens were used to generate polyclonal antibodies, which were screened for their ability to recognize orthologous proteins in P. knowlesi. These antibodies were then tested for inhibition of growth and invasion of both wild type P. knowlesi and chimeric P. knowlesi lines modified using CRISPR/Cas9 to exchange P. knowlesi genes with their P. vivax orthologues. Candidates that induced antibodies that inhibited invasion to a similar level as PvDBP were identified, confirming the utility of P. knowlesi as a model for P. vivax vaccine development and prioritizing antigens for further follow up., Author summary Malaria parasites cause disease after invading human red blood cells, implying that a vaccine that interrupts this process could play a significant role in malaria control. Multiple Plasmodium parasite species can cause malaria in humans, and most malaria outside Africa is caused by Plasmodium vivax. There is currently no effective vaccine against the blood stage of any malaria parasite, and progress in P. vivax vaccine development has been particularly hampered because this parasite species cannot be cultured for prolonged periods of time in the lab. We explored whether a related species, P. knowlesi, which can be propagated in human red blood cells in vitro, can be used to screen for potential P. vivax vaccine targets. We raised antibodies against selected P. vivax proteins and tested their ability to recognize and prevent P. knowlesi parasites from invading human red blood cells, thereby identifying multiple novel vaccine candidates.
- Published
- 2021
47. STEM CELL TECHNOLOGY PROVIDES NOVEL TOOLS TO UNDERSTAND HUMAN VARIATION IN Plasmodium falciparum MALARIA
- Author
-
Koutsourakis M, Ruddy Montandon, Ling B, Mwikali K, Alena Pance, Frances Law, Foad J. Rouhani, Julian C. Rayner, Hannes Ponstingl, and Chukwuma A. Agu
- Subjects
biology ,Basigin ,medicine ,Plasmodium falciparum ,Stem cell ,biology.organism_classification ,Induced pluripotent stem cell ,Receptor ,medicine.disease ,Reprogramming ,Gene ,Malaria ,Cell biology - Abstract
Plasmodium falciparum interacts with several human cell types during their complex life cycle, including erythrocytes and hepatocytes. The enuclated nature of erythrocytes makes them inaccessible to genetic tools, which in turn makes studying erythrocyte proteins involved in malaria invasion and development particularly difficult. Here we overcome this limitation using stem cell technology to develop a universal differentiation protocol for in vitro derivation of erythrocytes from a variety of stem cell lines of diverse origin. This allows manipulation of erythrocytic genes and examination of their impact on the parasite by flow cytometric detection of parasite haemozoin. Deletion of Basigin, the essential receptor for P. falciparum, abrogates invasion, while other less studied proteins such as ATP2B4 have a minor effect. Reprogramming of induced pluripotent stem cells from α-thalassemia primary samples shows reduced infection levels, demonstrating this approach is useful for understanding the effect of natural human polymorphisms on the disease.
- Published
- 2021
48. An open dataset of
- Author
-
Ambroise, Ahouidi, Mozam, Ali, Jacob, Almagro-Garcia, Alfred, Amambua-Ngwa, Chanaki, Amaratunga, Roberto, Amato, Lucas, Amenga-Etego, Ben, Andagalu, Tim J C, Anderson, Voahangy, Andrianaranjaka, Tobias, Apinjoh, Cristina, Ariani, Elizabeth A, Ashley, Sarah, Auburn, Gordon A, Awandare, Hampate, Ba, Vito, Baraka, Alyssa E, Barry, Philip, Bejon, Gwladys I, Bertin, Maciej F, Boni, Steffen, Borrmann, Teun, Bousema, Oralee, Branch, Peter C, Bull, George B J, Busby, Thanat, Chookajorn, Kesinee, Chotivanich, Antoine, Claessens, David, Conway, Alister, Craig, Umberto, D'Alessandro, Souleymane, Dama, Nicholas Pj, Day, Brigitte, Denis, Mahamadou, Diakite, Abdoulaye, Djimdé, Christiane, Dolecek, Arjen M, Dondorp, Chris, Drakeley, Eleanor, Drury, Patrick, Duffy, Diego F, Echeverry, Thomas G, Egwang, Berhanu, Erko, Rick M, Fairhurst, Abdul, Faiz, Caterina A, Fanello, Mark M, Fukuda, Dionicia, Gamboa, Anita, Ghansah, Lemu, Golassa, Sonia, Goncalves, William L, Hamilton, G L Abby, Harrison, Lee, Hart, Christa, Henrichs, Tran Tinh, Hien, Catherine A, Hill, Abraham, Hodgson, Christina, Hubbart, Mallika, Imwong, Deus S, Ishengoma, Scott A, Jackson, Chris G, Jacob, Ben, Jeffery, Anna E, Jeffreys, Kimberly J, Johnson, Dushyanth, Jyothi, Claire, Kamaliddin, Edwin, Kamau, Mihir, Kekre, Krzysztof, Kluczynski, Theerarat, Kochakarn, Abibatou, Konaté, Dominic P, Kwiatkowski, Myat Phone, Kyaw, Pharath, Lim, Chanthap, Lon, Kovana M, Loua, Oumou, Maïga-Ascofaré, Cinzia, Malangone, Magnus, Manske, Jutta, Marfurt, Kevin, Marsh, Mayfong, Mayxay, Alistair, Miles, Olivo, Miotto, Victor, Mobegi, Olugbenga A, Mokuolu, Jacqui, Montgomery, Ivo, Mueller, Paul N, Newton, Thuy, Nguyen, Thuy-Nhien, Nguyen, Harald, Noedl, Francois, Nosten, Rintis, Noviyanti, Alexis, Nzila, Lynette I, Ochola-Oyier, Harold, Ocholla, Abraham, Oduro, Irene, Omedo, Marie A, Onyamboko, Jean-Bosco, Ouedraogo, Kolapo, Oyebola, Richard D, Pearson, Norbert, Peshu, Aung Pyae, Phyo, Chris V, Plowe, Ric N, Price, Sasithon, Pukrittayakamee, Milijaona, Randrianarivelojosia, Julian C, Rayner, Pascal, Ringwald, Kirk A, Rockett, Katherine, Rowlands, Lastenia, Ruiz, David, Saunders, Alex, Shayo, Peter, Siba, Victoria J, Simpson, Jim, Stalker, Xin-Zhuan, Su, Colin, Sutherland, Shannon, Takala-Harrison, Livingstone, Tavul, Vandana, Thathy, Antoinette, Tshefu, Federica, Verra, Joseph, Vinetz, Thomas E, Wellems, Jason, Wendler, Nicholas J, White, Ian, Wright, William, Yavo, and Htut, Ye
- Subjects
data resource ,drug resistance ,plasmodium falciparum ,parasitic diseases ,evolution ,malaria ,genomics ,rapid diagnostic test failure ,population genetics ,Articles ,genomic epidemiology ,Research Article - Abstract
MalariaGEN is a data-sharing network that enables groups around the world to work together on the genomic epidemiology of malaria. Here we describe a new release of curated genome variation data on 7,000 Plasmodium falciparum samples from MalariaGEN partner studies in 28 malaria-endemic countries. High-quality genotype calls on 3 million single nucleotide polymorphisms (SNPs) and short indels were produced using a standardised analysis pipeline. Copy number variants associated with drug resistance and structural variants that cause failure of rapid diagnostic tests were also analysed. Almost all samples showed genetic evidence of resistance to at least one antimalarial drug, and some samples from Southeast Asia carried markers of resistance to six commonly-used drugs. Genes expressed during the mosquito stage of the parasite life-cycle are prominent among loci that show strong geographic differentiation. By continuing to enlarge this open data resource we aim to facilitate research into the evolutionary processes affecting malaria control and to accelerate development of the surveillance toolkit required for malaria elimination.
- Published
- 2021
49. The Plasmodium falciparum Rh5 invasion protein complex reveals an excess of rare variant mutations
- Author
-
George Githinji, Kevin Omondi Ochwedo, Julian C. Rayner, Lynette Isabella Ochola-Oyier, Philip Bejon, Victor Osoti, Kevin Wamae, Leonard Ndwiga, Wamae, Kevin [0000-0001-7721-5534], Ochola-Oyier, Lynette Isabella [0000-0003-4393-0470], and Apollo - University of Cambridge Repository
- Subjects
0301 basic medicine ,Linkage disequilibrium ,Single Nucleotide Polymorphisms ,RC955-962 ,Plasmodium falciparum ,Protozoan Proteins ,Single-nucleotide polymorphism ,Infectious and parasitic diseases ,RC109-216 ,Biology ,03 medical and health sciences ,0302 clinical medicine ,Rh5 ,Polymorphism (computer science) ,Arctic medicine. Tropical medicine ,Ripr ,Indel ,Gene ,Whole genome sequencing ,Genetics ,Polymorphism, Genetic ,Research ,biology.organism_classification ,3. Good health ,Malaria ,Minor allele frequency ,030104 developmental biology ,Infectious Diseases ,Cyrpa ,Multigene Family ,Mutation ,Parasitology ,P113 ,Carrier Proteins ,Vaccine ,linkage disequilibrium ,030215 immunology - Abstract
Background The invasion of the red blood cells by Plasmodium falciparum merozoites involves the interplay of several proteins that are also targets for vaccine development. The proteins PfRh5-PfRipr-PfCyRPA-Pfp113 assemble into a complex at the apical end of the merozoite and are together essential for erythrocyte invasion. They have also been shown to induce neutralizing antibodies and appear to be less polymorphic than other invasion-associated proteins, making them high priority blood-stage vaccine candidates. Using available whole genome sequencing data (WGS) and new capillary sequencing data (CS), this study describes the genetic polymorphism in the Rh5 complex in P. falciparum isolates obtained from Kilifi, Kenya. Methods 162 samples collected in 2013 and 2014 were genotyped by capillary sequencing (CS) and re-analysed WGS from 68 culture-adapted P. falciparum samples obtained from a drug trial conducted from 2005 to 2007. The frequency of polymorphisms in the merozoite invasion proteins, PfRh5, PfRipr, PfCyRPA and PfP113 were examined and where possible polymorphisms co-occurring in the same isolates. Results From a total 70 variants, including 2 indels, 19 SNPs [27.1%] were identified by both CS and WGS, while an additional 15 [21.4%] and 36 [51.4%] SNPs were identified only by either CS or WGS, respectively. All the SNPs identified by CS were non-synonymous, whereas WGS identified 8 synonymous and 47 non-synonymous SNPs. CS identified indels in repeat regions in the p113 gene in codons 275 and 859 that were not identified in the WGS data. The minor allele frequencies of the SNPs ranged between 0.7 and 34.9% for WGS and 1.1–29.6% for CS. Collectively, 12 high frequency SNPs (> 5%) were identified: four in Rh5 codon 147, 148, 203 and 429, two in p113 at codons 7 and 267 and six in Ripr codons 190, 259, 524, 985, 1003 and 1039. Conclusion This study reveals that the majority of the polymorphisms are rare variants and confirms a low level of genetic polymorphisms in all proteins within the Rh5 complex.
- Published
- 2021
50. Adaptation of Plasmodium falciparum to humans involved the loss of an ape-specific erythrocyte invasion ligand
- Author
-
Manoj T. Duraisingh, Alison Kemp, Julian C. Rayner, Selasi Dankwa, William R. Proto, Paul M. Sharp, Gavin J. Wright, Zenon A. Zenonos, Steve Unwin, Weimin Liu, Sasha V. Siegel, Sarah Marsden, Beatrice H. Hahn, Proto, William R [0000-0003-2311-920X], Siegel, Sasha V [0000-0002-1458-4348], Dankwa, Selasi [0000-0002-1837-2924], Sharp, Paul M [0000-0001-9771-543X], Hahn, Beatrice H [0000-0002-9400-9887], Rayner, Julian C [0000-0002-9835-1014], Apollo - University of Cambridge Repository, Proto, William R. [0000-0003-2311-920X], Siegel, Sasha V. [0000-0002-1458-4348], Sharp, Paul M. [0000-0001-9771-543X], Hahn, Beatrice H. [0000-0002-9400-9887], and Rayner, Julian C. [0000-0002-9835-1014]
- Subjects
0301 basic medicine ,Erythrocytes ,Protozoan Proteins ,General Physics and Astronomy ,medicine.disease_cause ,13 ,law.invention ,0302 clinical medicine ,law ,Loss of Function Mutation ,631/326/417/1716 ,Malaria, Falciparum ,lcsh:Science ,Frameshift Mutation ,Cell Engineering ,Genetics ,Gene Editing ,Mutation ,Multidisciplinary ,biology ,article ,Ligand (biochemistry) ,3. Good health ,Parasite biology ,13/31 ,Parasite evolution ,Recombinant DNA ,Pan troglodytes ,Science ,Plasmodium falciparum ,13/106 ,General Biochemistry, Genetics and Molecular Biology ,Laverania ,Host Specificity ,Frameshift mutation ,38/91 ,82/80 ,Evolution, Molecular ,03 medical and health sciences ,parasitic diseases ,medicine ,692/699/255/1629 ,Animals ,Humans ,631/326/417/2548 ,Gene ,HEK 293 cells ,General Chemistry ,biology.organism_classification ,Malaria ,030104 developmental biology ,HEK293 Cells ,Sialic Acids ,lcsh:Q ,CRISPR-Cas Systems ,030217 neurology & neurosurgery - Abstract
Plasmodium species are frequently host-specific, but little is currently known about the molecular factors restricting host switching. This is particularly relevant for P. falciparum, the only known human-infective species of the Laverania sub-genus, all other members of which infect African apes. Here we show that all tested P. falciparum isolates contain an inactivating mutation in an erythrocyte invasion associated gene, PfEBA165, the homologues of which are intact in all ape-infective Laverania species. Recombinant EBA165 proteins only bind ape, not human, erythrocytes, and this specificity is due to differences in erythrocyte surface sialic acids. Correction of PfEBA165 inactivating mutations by genome editing yields viable parasites, but is associated with down regulation of both PfEBA165 and an adjacent invasion ligand, which suggests that PfEBA165 expression is incompatible with parasite growth in human erythrocytes. Pseudogenization of PfEBA165 may represent a key step in the emergence and evolution of P. falciparum., Here, Proto et al. show that human infective Plasmodium falciparum isolates contain an inactivating mutation in the erythrocyte invasion associated gene PfEBA165, while homologues of ape-infective Laverania species are intact, and that expression of intact PfEBA165 is incompatible with parasite growth in human erythrocytes.
- Published
- 2019
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.