1
|
Little TS, Cunningham DA, Christophides GK, Reid AJ, Langhorne J. De novo assembly of plasmodium interspersed repeat (pir) genes from Plasmodium vivax RNAseq data suggests geographic conservation of sub-family transcription. BMC Genomics 2025; 26:544. [PMID: 40442603 PMCID: PMC12121038 DOI: 10.1186/s12864-025-11752-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 05/26/2025] [Indexed: 06/02/2025] Open
Abstract
BACKGROUND The plasmodium interspersed repeats (pir) multigene family is found across malaria parasite genomes, first discovered in the human-infecting species Plasmodium vivax, where they were initially named the virs. Their function remains unknown, although studies have suggested a role in virulence of the asexual blood stages. Sub-families of the P. vivax pir/virs have been identified, and are found in isolates from across the world, however their transcription at different localities and in different stages of the life cycle have not been quantified. Multiple transcriptomic studies of the parasite have been conducted, but many map the pir reads to existing reference genomes (as part of standard bioinformatic practice), which may miss members of the multigene family due to its inherent variability. This obscures our understanding of how the pir sub-families in P. vivax may be contributing to human/vector infection. RESULTS To overcome the issue of hidden pir diversity from utilising a reference genome, we employed de novo transcriptome assembly to construct the pir 'reference' of different parasite isolates from published and novel RNAseq datasets. For this purpose, a pipeline was written in Nextflow, and first tested on data from the rodent-infecting P. c. chabaudi parasite to ascertain its efficacy on a sample with a full, genome-based set of pir gene sequences. The pipeline assembled hundreds of pirs from the studies included. By performing BLAST sequence identity comparisons with reference genome pirs (including P. vivax and related species) we found a clustered network of transcripts which corresponded well with prior sub-family annotations, albeit requiring some updated nomenclature. Mapping the RNAseq datasets to the de novo transcriptome references revealed that the transcription of these updated pir gene sub-families is generally consistent across the different geographical regions. From this transcriptional quantification, a time course of mosquito bloodmeals (after feeding on an infected patient) highlighted the first evidence of ookinete stage pir transcription in a human-infective malaria parasite. CONCLUSIONS De novo transcriptome assembly is a valuable tool for understanding highly variable multigene families from Plasmodium spp., and with pipeline software these can be applied more easily and at scale. Despite a global distribution, P. vivax has a conserved pir sub-family structure-both in terms of genome copy number and transcription. We suggest that this indicates important roles of the distinct sub-families, or a genetic mechanism maintaining their preservation. Furthermore, a burst of pir transcription in the mosquito stages of development is the first glint of ookinete pir expression for a human-infective malaria parasite, suggesting a role for the gene family at a new stage of the lifecycle.
Collapse
Affiliation(s)
- Timothy S Little
- The Francis Crick Institute, Midland Road, London, UK
- Department of Life Sciences, Imperial College London, South Kensington, London, UK
- Present Address: UCL Respiratory, Rayne Building, University College London, London, UK
| | | | | | - Adam James Reid
- The Gurdon Institute, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QN, UK
| | | |
Collapse
|
2
|
Zadow ME, MacRaild CA, Creek DJ, Wilson DW. Alba protein-mediated gene and protein regulation in protozoan parasites. Int J Parasitol 2025:S0020-7519(25)00076-1. [PMID: 40246164 DOI: 10.1016/j.ijpara.2025.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 02/21/2025] [Accepted: 04/10/2025] [Indexed: 04/19/2025]
Abstract
The success of protozoan parasites relies heavily on regulation of gene and protein expression to facilitate their persistence in harsh and often changing environments. These parasites display biology that is highly divergent from model eukaryotes, unfortunately leaving our understanding of these parasites' critical regulatory mechanisms incomplete. Alba proteins, a highly diverse group of DNA/RNA-binding proteins, are found across all domains of life and it has become increasingly apparent that these proteins play key regulatory roles in many protozoan parasite species including Plasmodium, Leishmania, Toxoplasma, and Trypanosoma. This review focusses on a subset of clinically relevant protozoan parasites and highlights the key biological processes known to have Alba protein involvement in these organisms including parasite development, survival, and virulence. In order to gain greater insight into these proteins, we also undertook a bioinformatic exploration of their protein sequences, leading us to identify previously unreported C-terminal Alba domain motifs and propose annotations for several currently unannotated protozoan Alba-like proteins. This collation of information allows us to observe common themes in Alba protein function across this group of parasites while also identifying areas of opportunity for further study.
Collapse
Affiliation(s)
- Meghan E Zadow
- Research Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide 5005, Australia; Institute for Photonics and Advanced Sensing (IPAS), University of Adelaide 5005 SA, Australia.
| | - Christopher A MacRaild
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Darren J Creek
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Danny W Wilson
- Research Centre for Infectious Diseases, School of Biological Sciences, University of Adelaide, Adelaide 5005, Australia; Institute for Photonics and Advanced Sensing (IPAS), University of Adelaide 5005 SA, Australia; Burnet Institute, Melbourne 3004 Victoria, Australia.
| |
Collapse
|
3
|
Bento I, Parrington BA, Pascual R, Goldberg AS, Wang E, Liu H, Borrmann H, Zelle M, Coburn N, Takahashi JS, Elias JE, Mota MM, Rijo-Ferreira F. Parasite and vector circadian clocks mediate efficient malaria transmission. Nat Microbiol 2025; 10:882-896. [PMID: 40164831 PMCID: PMC11964930 DOI: 10.1038/s41564-025-01949-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 01/08/2025] [Indexed: 04/02/2025]
Abstract
Malaria transmission begins when Anopheles mosquitos deposit saliva and Plasmodium parasites during a bloodmeal. As Anopheles mosquitos are nocturnal, we investigated whether their salivary glands are under circadian control, anticipating bloodmeals and modulating parasite biology for host encounters. Here we show that approximately half of the mosquito salivary gland transcriptome, particularly genes essential for efficient bloodmeals such as anti-blood clotting factors, exhibits circadian expression. Furthermore, measuring haemoglobin levels, we demonstrate that mosquitos prefer to feed and ingest more blood at nighttime. Notably, we show a substantial subset of the salivary-gland-resident parasite transcriptome cycling throughout the day, indicating that this stage is not transcriptionally quiescent. Among the sporozoite genes undergoing rhythmic expression are those involved in parasite motility, potentially modulating the ability to initiate infection at different times of day. Our findings suggest a circadian tripartite relationship between the vector, parasite and mammalian host that together modulates malaria transmission.
Collapse
Affiliation(s)
- Inês Bento
- Gulbenkian Institute for Molecular Medicine, Lisbon, Portugal
| | - Brianna A Parrington
- Berkeley Public Health, Molecular and Cell Biology Department, University of California Berkeley, Berkeley, CA, USA
| | - Rushlenne Pascual
- Berkeley Public Health, Molecular and Cell Biology Department, University of California Berkeley, Berkeley, CA, USA
| | - Alexander S Goldberg
- Berkeley Public Health, Molecular and Cell Biology Department, University of California Berkeley, Berkeley, CA, USA
| | - Eileen Wang
- Chan Zuckerberg Biohub - San Francisco, San Francisco, CA, USA
| | - Hani Liu
- Berkeley Public Health, Molecular and Cell Biology Department, University of California Berkeley, Berkeley, CA, USA
| | - Helene Borrmann
- Berkeley Public Health, Molecular and Cell Biology Department, University of California Berkeley, Berkeley, CA, USA
| | - Mira Zelle
- Berkeley Public Health, Molecular and Cell Biology Department, University of California Berkeley, Berkeley, CA, USA
| | - Nicholas Coburn
- Berkeley Public Health, Molecular and Cell Biology Department, University of California Berkeley, Berkeley, CA, USA
| | - Joseph S Takahashi
- Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center Dallas, Dallas, TX, USA
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center Dallas, Dallas, TX, USA
| | - Joshua E Elias
- Chan Zuckerberg Biohub - San Francisco, San Francisco, CA, USA
| | - Maria M Mota
- Gulbenkian Institute for Molecular Medicine, Lisbon, Portugal
- Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Filipa Rijo-Ferreira
- Berkeley Public Health, Molecular and Cell Biology Department, University of California Berkeley, Berkeley, CA, USA.
- Chan Zuckerberg Biohub - San Francisco, San Francisco, CA, USA.
| |
Collapse
|
4
|
Saab SA, Cardoso-Jaime V, Kefi M, Dimopoulos G. Advances in the dissection of Anopheles-Plasmodium interactions. PLoS Pathog 2025; 21:e1012965. [PMID: 40163471 PMCID: PMC11957333 DOI: 10.1371/journal.ppat.1012965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025] Open
Abstract
Malaria is a life-threatening mosquito-borne disease caused by the Plasmodium parasite, responsible for more than half a million deaths annually and principally involving children. The successful transmission of malaria by Anopheles mosquitoes relies on complex successive interactions between the parasite and various mosquito organs, host factors, and restriction factors. This review summarizes our current understanding of the mechanisms regulating Plasmodium infection of the mosquito vector at successive plasmodial developmental stages and highlights potential transmission-blocking targets and strategies.
Collapse
Affiliation(s)
- Sally A. Saab
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States America
| | - Victor Cardoso-Jaime
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States America
| | - Mary Kefi
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States America
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States America
| |
Collapse
|
5
|
Rosado-Quiñones AM, Colón-Lorenzo EE, Pala ZR, Bosch J, Kudyba K, Kudyba H, Leed SE, Roncal N, Baerga-Ortiz A, Roche-Lima A, Gerena Y, Fidock DA, Roth A, Vega-Rodríguez J, Serrano AE. Novel hydrazone compounds with broad-spectrum antiplasmodial activity and synergistic interactions with antimalarial drugs. Antimicrob Agents Chemother 2024; 68:e0164323. [PMID: 38639491 PMCID: PMC11620517 DOI: 10.1128/aac.01643-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 03/20/2024] [Indexed: 04/20/2024] Open
Abstract
The development of novel antiplasmodial compounds with broad-spectrum activity against different stages of Plasmodium parasites is crucial to prevent malaria disease and parasite transmission. This study evaluated the antiplasmodial activity of seven novel hydrazone compounds (referred to as CB compounds: CB-27, CB-41, CB-50, CB-53, CB-58, CB-59, and CB-61) against multiple stages of Plasmodium parasites. All CB compounds inhibited blood stage proliferation of drug-resistant or sensitive strains of Plasmodium falciparum in the low micromolar to nanomolar range. Interestingly, CB-41 exhibited prophylactic activity against hypnozoites and liver schizonts in Plasmodium cynomolgi, a primate model for Plasmodium vivax. Four CB compounds (CB-27, CB-41, CB-53, and CB-61) inhibited P. falciparum oocyst formation in mosquitoes, and five CB compounds (CB-27, CB-41, CB-53, CB-58, and CB-61) hindered the in vitro development of Plasmodium berghei ookinetes. The CB compounds did not inhibit the activation of P. berghei female and male gametocytes in vitro. Isobologram assays demonstrated synergistic interactions between CB-61 and the FDA-approved antimalarial drugs, clindamycin and halofantrine. Testing of six CB compounds showed no inhibition of Plasmodium glutathione S-transferase as a putative target and no cytotoxicity in HepG2 liver cells. CB compounds are promising candidates for further development as antimalarial drugs against multidrug-resistant parasites, which could also prevent malaria transmission.
Collapse
Affiliation(s)
- Angélica M. Rosado-Quiñones
- Department of Microbiology and Medical Zoology, University of Puerto Rico School of Medicine, San Juan, Puerto Rico
| | - Emilee E. Colón-Lorenzo
- Department of Microbiology and Medical Zoology, University of Puerto Rico School of Medicine, San Juan, Puerto Rico
| | - Zarna Rajeshkumar Pala
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Jürgen Bosch
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, Ohio, USA
- InterRayBio, LLC, Cleveland, Ohio, USA
| | - Karl Kudyba
- Department of Drug Discovery, Experimental Therapeutics Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Heather Kudyba
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Susan E. Leed
- Department of Drug Discovery, Experimental Therapeutics Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Norma Roncal
- Department of Drug Discovery, Experimental Therapeutics Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Abel Baerga-Ortiz
- Department of Biochemistry, University of Puerto Rico School of Medicine, San Juan, Puerto Rico
| | - Abiel Roche-Lima
- RCMI Program, Medical Science Campus, University of Puerto Rico, San Juan, Puerto Rico
| | - Yamil Gerena
- Department of Pharmacology and Toxicology, University of Puerto Rico School of Medicine, San Juan, Puerto Rico
| | - David A. Fidock
- Department of Microbiology and Immunology, Columbia University, New York, New York, USA
- Division of Infectious Diseases, Department of Medicine, Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Medical Center, New York, New York, USA
| | - Alison Roth
- Department of Drug Discovery, Experimental Therapeutics Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Joel Vega-Rodríguez
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Adelfa E. Serrano
- Department of Microbiology and Medical Zoology, University of Puerto Rico School of Medicine, San Juan, Puerto Rico
| |
Collapse
|
6
|
Bento I, Parrington B, Pascual R, Goldberg AS, Wang E, Liu H, Zelle M, Takahashi JS, Elias JE, Mota MM, Rijo-Ferreira F. Circadian rhythms mediate malaria transmission potential. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.14.594221. [PMID: 38798622 PMCID: PMC11118478 DOI: 10.1101/2024.05.14.594221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Malaria transmission begins when infected female Anopheles mosquitos deposit Plasmodium parasites into the mammalian host's skin during a bloodmeal. The salivary gland-resident sporozoite parasites migrate to the bloodstream, subsequently invading and replicating within hepatocytes. As Anopheles mosquitos are more active at night, with a 24-hour rhythm, we investigated whether their salivary glands are under circadian control, anticipating bloodmeals and modulating sporozoite biology for host encounters. Here we show that approximately half of the mosquito salivary gland transcriptome, particularly genes essential for efficient bloodmeals such as anti-blood clotting factors, exhibits circadian rhythmic expression. Furthermore, we demonstrate that mosquitoes prefer to feed during nighttime, with the amount of blood ingested varying cyclically throughout the day. Notably, we show a substantial subset of the sporozoite transcriptome cycling throughout the day. These include genes involved in parasite motility, potentially modulating the ability to initiate infection at different times of day. Thus, although sporozoites are typically considered quiescent, our results demonstrate their transcriptional activity, revealing robust daily rhythms of gene expression. Our findings suggest a circadian evolutionary relationship between the vector, parasite and mammalian host that together modulate malaria transmission.
Collapse
|
7
|
Plata-Pineda SE, Cárdenas-Munévar LX, Castro-Cavadía CJ, Buitrago SP, Garzón-Ospina D. Evaluating the genetic diversity of the Plasmodium vivax siap2 locus: A promising candidate for an effective malaria vaccine? Acta Trop 2024; 251:107111. [PMID: 38151069 DOI: 10.1016/j.actatropica.2023.107111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 11/27/2023] [Accepted: 12/24/2023] [Indexed: 12/29/2023]
Abstract
Malaria is the deadliest parasitic disease in the world. Traditional control measures have become less effective; hence, there is a need to explore alternative strategies, such as antimalarial vaccines. However, designing an anti-Plasmodium vivax vaccine is considered a challenge due to the complex parasite biology and the antigens' high genetic diversity. Recently, the sporozoite invasion-associated protein 2 (SIAP2) has been suggested as a potential antigen to be considered in vaccine design due to its significance during hepatocyte invasion. However, its use may be limited by the incomplete understanding of gene/protein diversity. Here, the genetic diversity of pvsiap2 using P. vivax DNA samples from Colombia was assessed. Through PCR amplification and sequencing, we compared the Colombian sequences with available worldwide sequences, revealing that pvsiap2 displays low genetic diversity. Molecular evolutionary analyses showed that pvsiap2 appears to be influenced by directional selection. Moreover, the haplotypes found differ by a few mutational steps and several of them were shared between different geographical areas. On the other hand, several conserved regions within PvSIAP2 were predicted as potential B-cell or T-cell epitopes. Considering these characteristics and its role in hepatocyte invasion, the PvSIAP2 protein emerges as a promising antigen to be considered in a multi-antigen-multi-stage (multivalent) fully effective vaccine against P. vivax malaria.
Collapse
Affiliation(s)
- Sergio E Plata-Pineda
- School of Biological Sciences, Grupo de Estudios en Genética y Biología Molecular (GEBIMOL), Universidad Pedagógica y Tecnológica de Colombia - UPTC, Tunja, Boyacá, Colombia
| | - Laura X Cárdenas-Munévar
- School of Biological Sciences, Grupo de Estudios en Genética y Biología Molecular (GEBIMOL), Universidad Pedagógica y Tecnológica de Colombia - UPTC, Tunja, Boyacá, Colombia
| | - Carlos J Castro-Cavadía
- Grupo de Investigaciones Microbiológicas y Biomédicas de Córdoba (GIMBIC), School of Health Sciences, Universidad de Córdoba, Montería, Córdoba, Colombia
| | - Sindy P Buitrago
- School of Biological Sciences, Grupo de Estudios en Genética y Biología Molecular (GEBIMOL), Universidad Pedagógica y Tecnológica de Colombia - UPTC, Tunja, Boyacá, Colombia; Population Genetics And Molecular Evolution (PGAME), Fundación Scient, Tunja, Boyacá, Colombia
| | - Diego Garzón-Ospina
- School of Biological Sciences, Grupo de Estudios en Genética y Biología Molecular (GEBIMOL), Universidad Pedagógica y Tecnológica de Colombia - UPTC, Tunja, Boyacá, Colombia; Population Genetics And Molecular Evolution (PGAME), Fundación Scient, Tunja, Boyacá, Colombia.
| |
Collapse
|
8
|
Ntumngia FB, Kolli SK, Annamalai Subramani P, Barnes SJ, Nicholas J, Ogbondah MM, Barnes BB, Salinas ND, Thawornpan P, Tolia NH, Chootong P, Adams JH. Naturally acquired antibodies against Plasmodium vivax pre-erythrocytic stage vaccine antigens inhibit sporozoite invasion of human hepatocytes in vitro. Sci Rep 2024; 14:1260. [PMID: 38218737 PMCID: PMC10787766 DOI: 10.1038/s41598-024-51820-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 01/09/2024] [Indexed: 01/15/2024] Open
Abstract
In Plasmodium vivax, the most studied vaccine antigens are aimed at blocking merozoite invasion of erythrocytes and disease development. Very few studies have evaluated pre-erythrocytic (PE) stage antigens. The P. vivax circumsporozoite protein (CSP), is considered the leading PE vaccine candidate, but immunity to CSP is short-lived and variant specific. Thus, there is a need to identify other potential candidates to partner with CSP in a multivalent vaccine to protect against infection and disease. We hypothesize that sporozoite antigens important for host cell infection are considered potential targets. In this study, we evaluated the magnitude and quality of naturally acquired antibody responses to four P. vivax PE antigens: sporozoite surface protein 3 (SSP3), sporozoite protein essential for traversal 1 (SPECT1), cell traversal protein of ookinetes and sporozoites (CelTOS) and CSP in plasma of P. vivax infected patients from Thailand. Naturally acquired antibodies to these antigens were prevalent in the study subjects, but with significant differences in magnitude of IgG antibody responses. About 80% of study participants had antibodies to all four antigens and only 2% did not have antibodies to any of the antigens. Most importantly, these antibodies inhibited sporozoite infection of hepatocytes in vitro. Significant variations in magnitude of antigen-specific inhibitory antibody responses were observed with individual samples. The highest inhibitory responses were observed with anti-CelTOS antibodies, followed by anti-SPECT1, SSP3 and CSP antibodies respectively. These data highlight the vaccine potential of these antigens in protecting against hepatocyte infection and the need for a multi-valent pre-erythrocytic vaccine to prevent liver stage development of P. vivax sporozoites.
Collapse
Affiliation(s)
- Francis Babila Ntumngia
- Center for Global Health and Interdisciplinary Research, University of South Florida, Tampa, FL, USA.
| | - Surendra Kumar Kolli
- Center for Global Health and Interdisciplinary Research, University of South Florida, Tampa, FL, USA
| | | | - Samantha J Barnes
- Center for Global Health and Interdisciplinary Research, University of South Florida, Tampa, FL, USA
| | - Justin Nicholas
- Center for Global Health and Interdisciplinary Research, University of South Florida, Tampa, FL, USA
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Madison M Ogbondah
- Center for Global Health and Interdisciplinary Research, University of South Florida, Tampa, FL, USA
| | - Brian B Barnes
- College of Marine Science, University of South Florida, St Petersburg, FL, USA
| | - Nichole D Salinas
- Host Pathogen Interactions and Structural Vaccinology Section, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Pongsakorn Thawornpan
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| | - Niraj H Tolia
- Host Pathogen Interactions and Structural Vaccinology Section, Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Patchanee Chootong
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| | - John H Adams
- Center for Global Health and Interdisciplinary Research, University of South Florida, Tampa, FL, USA.
| |
Collapse
|
9
|
Alvarez-Jarreta J, Amos B, Aurrecoechea C, Bah S, Barba M, Barreto A, Basenko EY, Belnap R, Blevins A, Böhme U, Brestelli J, Brown S, Callan D, Campbell LI, Christophides GK, Crouch K, Davison HR, DeBarry JD, Demko R, Doherty R, Duan Y, Dundore W, Dyer S, Falke D, Fischer S, Gajria B, Galdi D, Giraldo-Calderón GI, Harb OS, Harper E, Helb D, Howington C, Hu S, Humphrey J, Iodice J, Jones A, Judkins J, Kelly SA, Kissinger JC, Kittur N, Kwon DK, Lamoureux K, Li W, Lodha D, MacCallum RM, Maslen G, McDowell MA, Myers J, Nural MV, Roos DS, Rund SSC, Shanmugasundram A, Sitnik V, Spruill D, Starns D, Tomko SS, Wang H, Warrenfeltz S, Wieck R, Wilkinson PA, Zheng J. VEuPathDB: the eukaryotic pathogen, vector and host bioinformatics resource center in 2023. Nucleic Acids Res 2024; 52:D808-D816. [PMID: 37953350 PMCID: PMC10767879 DOI: 10.1093/nar/gkad1003] [Citation(s) in RCA: 78] [Impact Index Per Article: 78.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/09/2023] [Accepted: 10/19/2023] [Indexed: 11/14/2023] Open
Abstract
The Eukaryotic Pathogen, Vector and Host Informatics Resource (VEuPathDB, https://veupathdb.org) is a Bioinformatics Resource Center funded by the National Institutes of Health with additional funding from the Wellcome Trust. VEuPathDB supports >600 organisms that comprise invertebrate vectors, eukaryotic pathogens (protists and fungi) and relevant free-living or non-pathogenic species or hosts. Since 2004, VEuPathDB has analyzed omics data from the public domain using contemporary bioinformatic workflows, including orthology predictions via OrthoMCL, and integrated the analysis results with analysis tools, visualizations, and advanced search capabilities. The unique data mining platform coupled with >3000 pre-analyzed data sets facilitates the exploration of pertinent omics data in support of hypothesis driven research. Comparisons are easily made across data sets, data types and organisms. A Galaxy workspace offers the opportunity for the analysis of private large-scale datasets and for porting to VEuPathDB for comparisons with integrated data. The MapVEu tool provides a platform for exploration of spatially resolved data such as vector surveillance and insecticide resistance monitoring. To address the growing body of omics data and advances in laboratory techniques, VEuPathDB has added several new data types, searches and features, improved the Galaxy workspace environment, redesigned the MapVEu interface and updated the infrastructure to accommodate these changes.
Collapse
Affiliation(s)
| | - Beatrice Amos
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | | | - Saikou Bah
- School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | | | - Ana Barreto
- University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Evelina Y Basenko
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | | | - Ann Blevins
- University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | | | | | - Stuart Brown
- University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | - Kathryn Crouch
- School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - Helen R Davison
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | | | - Richard Demko
- University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ryan Doherty
- University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yikun Duan
- University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Sarah Dyer
- European Bioinformatics Institute, Hinxton CB10 1SD, UK
| | - Dave Falke
- University of Georgia, Athens, GA 30602, USA
| | - Steve Fischer
- University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bindu Gajria
- University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daniel Galdi
- University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Omar S Harb
- University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Danica Helb
- University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Sufen Hu
- University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - John Iodice
- University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Andrew Jones
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | - John Judkins
- University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sarah A Kelly
- Imperial College London, South Kensington, London SW7 2BU, UK
| | | | | | - Dae Kun Kwon
- University of Notre Dame, Notre Dame, IN 46556, USA
| | | | - Wei Li
- University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Disha Lodha
- European Bioinformatics Institute, Hinxton CB10 1SD, UK
| | | | - Gareth Maslen
- Imperial College London, South Kensington, London SW7 2BU, UK
| | | | - Jeremy Myers
- University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - David S Roos
- University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Achchuthan Shanmugasundram
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
- Genomics England Limited, London E14 5AB, UK
| | - Vasily Sitnik
- European Bioinformatics Institute, Hinxton CB10 1SD, UK
| | | | - David Starns
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | | | | | | | - Robert Wieck
- University of Notre Dame, Notre Dame, IN 46556, USA
| | - Paul A Wilkinson
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | - Jie Zheng
- University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
10
|
Thawornpan P, Nicholas J, Malee C, Kochayoo P, Wangriatisak K, Tianpothong P, Ntumngia FB, J. Barnes S, H. Adams J, Chootong P. Longitudinal analysis of antibody responses to Plasmodium vivax sporozoite antigens following natural infection. PLoS Negl Trop Dis 2024; 18:e0011907. [PMID: 38277340 PMCID: PMC10817200 DOI: 10.1371/journal.pntd.0011907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 01/08/2024] [Indexed: 01/28/2024] Open
Abstract
BACKGROUND P. vivax malaria is a major global health burden hindering social and economic development throughout many tropical and sub-tropical countries. Pre-erythrocytic (PE) vaccines emerge as an attractive approach for the control and elimination of malaria infection. Therefore, evaluating the magnitude, longevity and prevalence of naturally acquired IgG antibody responses against PE candidate antigens is useful for vaccine design. METHODOLOGY/PRINCIPAL FINDINGS The antigenicity of five recombinant PE antigens (PvCSP-VK210, PvSSP3, PvM2-MAEBL, PvCelTOS and PvSPECT1) was evaluated in plasma samples from individuals residing in low transmission areas in Thailand (Ranong and Chumphon Provinces). The samples were collected at the time of acute vivax malaria and 90, 270 and 360 days later. The prevalence, magnitude and longevity of total IgG and IgG subclasses were determined for each antigen using the longitudinal data. Our results showed that seropositivity of all tested PE antigens was detected during infection in at least some subjects; anti-PvCSP-VK210 and anti-PvCelTOS antibodies were the most frequent. Titers of these antibodies declined during the year of follow up, but notably seropositivity persisted. Among seropositive subjects at post-infection, high number of subjects possessed antibodies against PvCSP-VK210. Anti-PvSSP3 antibody responses had the longest half-life. IgG subclass profiling showed that the predominant subclasses were IgG1 and IgG3 (cytophilic antibodies), tending to remain detectable for at least 360 days after infection. CONCLUSIONS/SIGNIFICANCE The present study demonstrated the magnitude and longevity of serological responses to multiple PE antigens of P. vivax after natural infection. This knowledge could contribute to the design of an effective P. vivax vaccine.
Collapse
Affiliation(s)
- Pongsakorn Thawornpan
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| | - Justin Nicholas
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, Tampa, Florida, United States of America
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, United States of America
| | - Chayapat Malee
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| | - Piyawan Kochayoo
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| | - Kittikorn Wangriatisak
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| | - Pachara Tianpothong
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| | - Francis Babila Ntumngia
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, Tampa, Florida, United States of America
| | - Samantha J. Barnes
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, Tampa, Florida, United States of America
| | - John H. Adams
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, Tampa, Florida, United States of America
| | - Patchanee Chootong
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| |
Collapse
|
11
|
Nicholas J, Kolli SK, Subramani PA, De SL, Ogbondah MM, Barnes SJ, Ntumngia FB, Adams JH. Comparative analyses of functional antibody-mediated inhibition with anti-circumsporozoite monoclonal antibodies against transgenic Plasmodium berghei. Malar J 2023; 22:335. [PMID: 37936181 PMCID: PMC10629016 DOI: 10.1186/s12936-023-04765-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 10/24/2023] [Indexed: 11/09/2023] Open
Abstract
BACKGROUND Acquired functional inhibitory antibodies are one of several humoral immune mechanisms used to neutralize foreign pathogens. In vitro bioassays are useful tools for quantifying antibody-mediated inhibition and evaluating anti-parasite immune antibodies. However, a gap remains in understanding of how antibody-mediated inhibition in vitro translates to inhibition in vivo. In this study, two well-characterized transgenic Plasmodium berghei parasite lines, PbmCh-luc and Pb-PfCSP(r), and murine monoclonal antibodies (mAbs) specific to P. berghei and Plasmodium falciparum circumsporozoite protein (CSP), 3D11 and 2A10, respectively, were used to evaluate antibody-mediated inhibition of parasite development in both in vitro and in vivo functional assays. METHODS IC50 values of mAbs were determined using an established inhibition of liver-stage development assay (ILSDA). For the in vivo inhibition assay, mice were passively immunized by transfer of the mAbs and subsequently challenged with 5.0 × 103 sporozoites via tail vein injection. The infection burden in both assays was quantified by luminescence and qRT-PCR of P. berghei 18S rRNA normalized to host GAPDH. RESULTS The IC50 values quantified by relative luminescence of mAbs 3D11 and 2A10 were 0.396 µg/ml and 0.093 µg/ml, respectively, against transgenic lines in vitro. Using the highest (> 90%) inhibitory antibody concentrations in a passive transfer, an IC50 of 233.8 µg/ml and 181.5 µg/ml for mAbs 3D11 and 2A10, respectively, was observed in vivo. At 25 µg (250 µg/ml), the 2A10 antibody significantly inhibited liver burden in mice compared to control. Additionally, qRT-PCR of P. berghei 18S rRNA served as a secondary validation of liver burden quantification. CONCLUSIONS Results from both experimental models, ILSDA and in vivo challenge, demonstrated that increased concentrations of the homologous anti-CSP repeat mAbs increased parasite inhibition. However, differences in antibody IC50 values between parasite lines did not allow a direct correlation between the inhibition of sporozoite invasion in vitro by ILSDA and the inhibition of mouse liver stage burden. Further studies are needed to establish the conditions for confident predictions for the in vitro ILSDA to be a predictor of in vivo outcomes using this model system.
Collapse
Affiliation(s)
- Justin Nicholas
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa, FL, 33612, USA
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Surendra Kumar Kolli
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa, FL, 33612, USA
| | - Pradeep Annamalai Subramani
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa, FL, 33612, USA
| | - Sai Lata De
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa, FL, 33612, USA
- Department of Infectious Disease & Immunology, College of Veterinary Medicine, University of Florida, Gainesville, FL, 32611, USA
| | - Madison M Ogbondah
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa, FL, 33612, USA
| | - Samantha J Barnes
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa, FL, 33612, USA
| | - Francis Babila Ntumngia
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa, FL, 33612, USA
| | - John H Adams
- Center for Global Health and Interdisciplinary Research, College of Public Health, University of South Florida, 3720 Spectrum Blvd, Tampa, FL, 33612, USA.
| |
Collapse
|
12
|
Ferraboli JW, Soares da Veiga GT, Albrecht L. Plasmodium vivax transcriptomics: What is new? Exp Biol Med (Maywood) 2023; 248:1645-1656. [PMID: 37786955 PMCID: PMC10723030 DOI: 10.1177/15353702231198070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2023] Open
Abstract
Malaria is the leading human parasitosis and is transmitted through the bite of anopheline mosquitoes infected with parasites of the genus Plasmodium spp. Among the seven species that cause malaria in humans, Plasmodium vivax is the most prevalent species in Latin America. In recent years, there have been an increasing number of reports of clinical complications caused by P. vivax infections, which were previously neglected and underestimated. P. vivax biology remains with large gaps. The emergence of next-generation sequencing technology has ensured a breakthrough in species knowledge. Coupled with this, the deposition of the P. vivax Sal-1 reference genome allowed an increase in transcriptomics projects by accessing messenger RNA. Thus, the regulation of differential gene expression according to the parasite life stage was verified, and several expressed genes were linked to different biological functions. Today, with the progress associated with RNA sequencing technologies, it is possible to detect nuances and obtain robust results. Discoveries provided by transcriptomic studies allow us to understand topics such as RNA expression and regulation and proteins and metabolic pathways involved during different stages of the parasite life cycle. The information obtained enables a better comprehension of immune system evasion mechanisms; invasion and adhesion strategies used by the parasite; as well as new vaccine targets, potential molecular markers, and others therapeutic targets. In this review, we provide new insights into P. vivax biology by summarizing recent findings in transcriptomic studies.
Collapse
Affiliation(s)
- Julia Weber Ferraboli
- Laboratory of Apicomplexan Parasites Research, Carlos Chagas Institute, Oswaldo Cruz Foundation (FIOCRUZ), Curitiba 81310-020, Brazil
| | - Gisele Tatiane Soares da Veiga
- Laboratory of Apicomplexan Parasites Research, Carlos Chagas Institute, Oswaldo Cruz Foundation (FIOCRUZ), Curitiba 81310-020, Brazil
| | - Letusa Albrecht
- Laboratory of Apicomplexan Parasites Research, Carlos Chagas Institute, Oswaldo Cruz Foundation (FIOCRUZ), Curitiba 81310-020, Brazil
| |
Collapse
|
13
|
Quaye IK, Aleksenko L, Paganotti GM, Peloewetse E, Haiyambo DH, Ntebela D, Oeuvray C, Greco B, the PAVON Consortium. Malaria Elimination in Africa: Rethinking Strategies for Plasmodium vivax and Lessons from Botswana. Trop Med Infect Dis 2023; 8:392. [PMID: 37624330 PMCID: PMC10458071 DOI: 10.3390/tropicalmed8080392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 07/21/2023] [Accepted: 07/22/2023] [Indexed: 08/26/2023] Open
Abstract
The global malaria community has picked up the theme of malaria elimination in more than 90% of the world's population in the next decade. Recent reports of Plasmodium vivax (P. vivax) in sub-Saharan Africa, including in Duffy-negative individuals, threaten the efforts aimed at achieving elimination. This is not only in view of strategies that are tailored only to P. falciparum elimination but also due to currently revealed biological characteristics of P. vivax concerning the relapse patterns of hypnozoites and conservation of large biomasses in cryptic sites in the bone marrow and spleen. A typical scenario was observed in Botswana between 2008 and 2018, which palpably projects how P. vivax could endanger malaria elimination efforts where the two parasites co-exist. The need for the global malaria community, national malaria programs (NMPs), funding agencies and relevant stakeholders to engage in a forum to discuss and recommend clear pathways for elimination of malaria, including P. vivax, in sub-Saharan Africa is warranted.
Collapse
Affiliation(s)
- Isaac K. Quaye
- Pan African Vivax and Ovale Network, Faculty of Engineering Computer and Allied Sciences, Regent University College of Science and Technology, #1 Regent Ave, McCarthy Hill, Mendskrom, Dansoman, Accra P.O. Box DS1636, Ghana
| | - Larysa Aleksenko
- Department of Health Sciences, School of Public Health, College of Health, Medicine and Life Sciences, Brunel University, Kingston Lane, Uxbridge, Middlesex, London UB8 3PH, UK;
| | - Giacomo M. Paganotti
- Botswana-University of Pennsylvania Partnership, Riverwalk, Gaborone P.O. Box 45498, Botswana;
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Elias Peloewetse
- Department of Biological Sciences, Faculty of Sciences, University of Botswana, Gaborone Private Bag 00704, Botswana;
| | - Daniel H. Haiyambo
- Department of Human, Biological and Translational Medical Sciences, Faculty of Health Sciences and Veterinary Medicine, University of Namibia School of Medicine, Hage Geingob Campus, Windhoek Private Bag 13301, Namibia;
| | - Davies Ntebela
- National Malaria Program, Ministry of Health, Gaborone Private Bag 0038, Botswana;
| | - Claude Oeuvray
- Global Health Institute of Merck, Terre Bonne Building Z0, Route de Crassier 1, Eysin, 1266 Geneva, Switzerland; (C.O.); (B.G.)
| | - Beatrice Greco
- Global Health Institute of Merck, Terre Bonne Building Z0, Route de Crassier 1, Eysin, 1266 Geneva, Switzerland; (C.O.); (B.G.)
| | - the PAVON Consortium
- PAVON, Regent University College of Science and Technology, #1 Regent Avenue, McCarthy Hiil, Mendskrom, Dansoman, Accra P.O. Box DS1636, Ghana
| |
Collapse
|
14
|
Avila-Bonilla R, Velazquez-Guzman J, Reyes-Zepeda E, Gutierrez-Avila J, Reyes-López C, Cisneros-Sarabia A, Saavedra E, Lopéz-Sandoval A, Ramírez-Moreno E, López-Camarillo C, Marchat L. Comparative genomics and interactomics of polyadenylation factors for the prediction of new parasite targets: Entamoeba histolytica as a working model. Biosci Rep 2023; 43:BSR20221911. [PMID: 36651565 PMCID: PMC9912109 DOI: 10.1042/bsr20221911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 01/05/2023] [Accepted: 01/13/2023] [Indexed: 01/19/2023] Open
Abstract
Protein-protein interactions (PPI) play a key role in predicting the function of a target protein and drug ability to affect an entire biological system. Prediction of PPI networks greatly contributes to determine a target protein and signal pathways related to its function. Polyadenylation of mRNA 3'-end is essential for gene expression regulation and several polyadenylation factors have been shown as valuable targets for controlling protozoan parasites that affect human health. Here, by using a computational strategy based on sequence-based prediction approaches, phylogenetic analyses, and computational prediction of PPI networks, we compared interactomes of polyadenylation factors in relevant protozoan parasites and the human host, to identify key proteins and define potential targets for pathogen control. Then, we used Entamoeba histolytica as a working model to validate our computational results. RT-qPCR assays confirmed the coordinated modulation of connected proteins in the PPI network and evidenced that silencing of the bottleneck protein EhCFIm25 affects the expression of interacting proteins. In addition, molecular dynamics simulations and docking approaches allowed to characterize the relationships between EhCFIm25 and Ehnopp34, two connected bottleneck proteins. Interestingly, the experimental identification of EhCFIm25 interactome confirmed the close relationships among proteins involved in gene expression regulation and evidenced new links with moonlight proteins in E. histolytica, suggesting a connection between RNA biology and metabolism as described in other organisms. Altogether, our results strengthened the relevance of comparative genomics and interactomics of polyadenylation factors for the prediction of new targets for the control of these human pathogens.
Collapse
Affiliation(s)
| | - Jorge Antonio Velazquez-Guzman
- Facultad de Ciencias, Universidad Autónoma del Estado de México. Carretera Toluca-Ixtlahuaca km 15.5 Cerrillo Piedras Blancas 50200 Toluca, Estado de México, Mexico
| | - Eimy Itzel Reyes-Zepeda
- Facultad de Ciencias, Universidad Autónoma del Estado de México. Carretera Toluca-Ixtlahuaca km 15.5 Cerrillo Piedras Blancas 50200 Toluca, Estado de México, Mexico
| | - Jorge Luis Gutierrez-Avila
- Posgrado en Ciencias Químico-Biológicas; Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional. Mexico City, Mexico
| | - César A Reyes-López
- Laboratorio de Bioquímica Estructural, Instituto Politécnico Nacional, Escuela Nacional de Medicina y Homeopatía, Mexico City 07320, Mexico
| | - Alondra Cisneros-Sarabia
- Laboratorio de Biomedicina Molecular II, ENMH, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Emma Saavedra
- Departamento de Bioquímica, Instituto Nacional de Cardiología, Mexico City 14080, Mexico
| | - Angel Lopéz-Sandoval
- Laboratorio de Biomedicina Molecular II, ENMH, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Esther Ramírez-Moreno
- Laboratorio de Biomedicina Molecular II, ENMH, Instituto Politécnico Nacional, Mexico City, Mexico
| | - César López-Camarillo
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México (UACM), Mexico City, Mexico
| | - Laurence A. Marchat
- Laboratorio de Biomedicina Molecular II, ENMH, Instituto Politécnico Nacional, Mexico City, Mexico
| |
Collapse
|
15
|
Mazhari R, Takashima E, Longley RJ, Ruybal-Pesantez S, White MT, Kanoi BN, Nagaoka H, Kiniboro B, Siba P, Tsuboi T, Mueller I. Identification of novel Plasmodium vivax proteins associated with protection against clinical malaria. Front Cell Infect Microbiol 2023; 13:1076150. [PMID: 36761894 PMCID: PMC9905245 DOI: 10.3389/fcimb.2023.1076150] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 01/09/2023] [Indexed: 01/26/2023] Open
Abstract
As progress towards malaria elimination continues, the challenge posed by the parasite species Plasmodium vivax has become more evident. In many regions co-endemic for P. vivax and Plasmodium falciparum, as transmission has declined the proportion of cases due to P. vivax has increased. Novel tools that directly target P. vivax are thus warranted for accelerated elimination. There is currently no advanced vaccine for P. vivax and only a limited number of potential candidates in the pipeline. In this study we aimed to identify promising P. vivax proteins that could be used as part of a subunit vaccination approach. We screened 342 P. vivax protein constructs for their ability to induce IgG antibody responses associated with protection from clinical disease in a cohort of children from Papua New Guinea. This approach has previously been used to successfully identify novel candidates. We were able to confirm previous results from our laboratory identifying the proteins reticulocyte binding protein 2b and StAR-related lipid transfer protein, as well as at least four novel candidates with similar levels of predicted protective efficacy. Assessment of these P. vivax proteins in further studies to confirm their potential and identify functional mechanisms of protection against clinical disease are warranted.
Collapse
Affiliation(s)
- Ramin Mazhari
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Eizo Takashima
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Ehime, Japan
| | - Rhea J Longley
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Shazia Ruybal-Pesantez
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Michael T White
- Institut Pasteur, Université de Paris Cité, G5 Épidémiologie et Analyse des Maladies Infectieuses, Département de Santé Globale, Paris, France
| | - Bernard N Kanoi
- Centre for Research in Infectious Diseases, Directorate of Research and Innovation, Mount Kenya University, Thika, Kenya
| | - Hikaru Nagaoka
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Ehime, Japan
| | - Benson Kiniboro
- Vector Borne Disease Unit, Papua New Guinea Institute of Medical Research, Goroka, Papua New Guinea
| | - Peter Siba
- Vector Borne Disease Unit, Papua New Guinea Institute of Medical Research, Goroka, Papua New Guinea
| | - Takafumi Tsuboi
- Division of Cell-Free Sciences, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Ivo Mueller
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
16
|
Bouyssou I, Martínez FJ, Campagne P, Ma L, Doderer-Lang C, Chitnis CE, Ménard D. Plasmodium vivax blood stage invasion pathways: Contribution of omics technologies in deciphering molecular and cellular mechanisms. C R Biol 2022; 345:91-133. [PMID: 36847467 DOI: 10.5802/crbiol.95] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 10/27/2022] [Indexed: 11/25/2022]
Abstract
Vivax malaria is an infectious disease caused by Plasmodium vivax, a parasitic protozoan transmitted by female Anopheline mosquitoes. Historically, vivax malaria has often been regarded as a benign self-limiting infection due to the observation of low parasitemia in Duffy-positive patients in endemic transmission areas and the virtual absence of infections in Duffy-negative individuals in Sub Saharan Africa. However, the latest estimates show that the burden of the disease is not decreasing in many countries and cases of vivax infections in Duffy-negative individuals are increasingly reported throughout Africa. This raised questions about the accuracy of diagnostics and the evolution of interactions between humans and parasites. For a long time, our knowledge on P. vivax biology has been hampered due to the limited access to biological material and the lack of robust in vitro culture methods. Consequently, little is currently known about P. vivax blood stage invasion mechanisms. The introduction of omics technologies with novel and accessible techniques such as third generation sequencing and RNA sequencing at single cell level, two-dimensional electrophoresis, liquid chromatography, and mass spectrometry, has progressively improved our understanding of P. vivax genetics, transcripts, and proteins. This review aims to provide broad insights into P. vivax invasion mechanisms generated by genomics, transcriptomics, and proteomics and to illustrate the importance of integrated multi-omics studies.
Collapse
|
17
|
Valenciano AL, Gomez-Lorenzo MG, Vega-Rodríguez J, Adams JH, Roth A. In vitro models for human malaria: targeting the liver stage. Trends Parasitol 2022; 38:758-774. [PMID: 35780012 PMCID: PMC9378454 DOI: 10.1016/j.pt.2022.05.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/27/2022] [Accepted: 05/27/2022] [Indexed: 11/16/2022]
Abstract
The Plasmodium liver stage represents a vulnerable therapeutic target to prevent disease progression as the parasite resides in the liver before clinical representation caused by intraerythrocytic development. However, most antimalarial drugs target the blood stage of the parasite's life cycle, and the few drugs that target the liver stage are lethal to patients with a glucose-6-phosphate dehydrogenase deficiency. Furthermore, implementation of in vitro liver models to study and develop novel therapeutics against the liver stage of human Plasmodium species remains challenging. In this review, we focus on the progression of in vitro liver models developed for human Plasmodium spp. parasites, provide a brief review on important assay requirements, and lastly present recommendations to improve models to enhance the discovery process of novel preclinical therapeutics.
Collapse
Affiliation(s)
- Ana Lisa Valenciano
- Center for Global Health and Infectious Diseases, College of Public Health, University of South Florida, Tampa, FL 33612, USA; Global Health Medicines R&D, GlaxoSmithKline, Severo Ochoa 2, Tres Cantos 28760, Madrid, Spain
| | - Maria G Gomez-Lorenzo
- Global Health Medicines R&D, GlaxoSmithKline, Severo Ochoa 2, Tres Cantos 28760, Madrid, Spain
| | - Joel Vega-Rodríguez
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - John H Adams
- Center for Global Health and Infectious Diseases, College of Public Health, University of South Florida, Tampa, FL 33612, USA
| | - Alison Roth
- Department of Drug Discovery, Experimental Therapeutics Branch, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA.
| |
Collapse
|
18
|
Gualdrón-López M, Díaz-Varela M, Zanghi G, Aparici-Herraiz I, Steel RW, Schäfer C, Cuscó P, Chuenchob V, Kangwangransan N, Billman ZP, Olsen TM, González JR, Roobsoong W, Sattabongkot J, Murphy SC, Mikolajczak SA, Borràs E, Sabidó E, Fernandez-Becerra C, Flannery EL, Kappe SH, del Portillo HA. Mass Spectrometry Identification of Biomarkers in Extracellular Vesicles From Plasmodium vivax Liver Hypnozoite Infections. Mol Cell Proteomics 2022; 21:100406. [PMID: 36030044 PMCID: PMC9520272 DOI: 10.1016/j.mcpro.2022.100406] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 08/12/2022] [Accepted: 08/20/2022] [Indexed: 01/18/2023] Open
Abstract
Latent liver stages termed hypnozoites cause relapsing Plasmodium vivax malaria infection and represent a major obstacle in the goal of malaria elimination. Hypnozoites are clinically undetectable, and presently, there are no biomarkers of this persistent parasite reservoir in the human liver. Here, we have identified parasite and human proteins associated with extracellular vesicles (EVs) secreted from in vivo infections exclusively containing hypnozoites. We used P. vivax-infected human liver-chimeric (huHEP) FRG KO mice treated with the schizonticidal experimental drug MMV048 as hypnozoite infection model. Immunofluorescence-based quantification of P. vivax liver forms showed that MMV048 removed schizonts from chimeric mice livers. Proteomic analysis of EVs derived from FRG huHEP mice showed that human EV cargo from infected FRG huHEP mice contain inflammation markers associated with active schizont replication and identified 66 P. vivax proteins. To identify hypnozoite-specific proteins associated with EVs, we mined the proteome data from MMV048-treated mice and performed an analysis involving intragroup and intergroup comparisons across all experimental conditions followed by a peptide compatibility analysis with predicted spectra to warrant robust identification. Only one protein fulfilled this stringent top-down selection, a putative filamin domain-containing protein. This study sets the stage to unveil biological features of human liver infections and identify biomarkers of hypnozoite infection associated with EVs.
Collapse
Affiliation(s)
- Melisa Gualdrón-López
- ISGlobal, Barcelona Institute for Global Health, Barcelona, Spain,IGTP, Institute for Health Sciences Trias I Pujol, Barcelona, Spain
| | - Miriam Díaz-Varela
- ISGlobal, Barcelona Institute for Global Health, Barcelona, Spain,IGTP, Institute for Health Sciences Trias I Pujol, Barcelona, Spain
| | - Gigliola Zanghi
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Iris Aparici-Herraiz
- ISGlobal, Barcelona Institute for Global Health, Barcelona, Spain,IGTP, Institute for Health Sciences Trias I Pujol, Barcelona, Spain
| | - Ryan W.J. Steel
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Carola Schäfer
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Pol Cuscó
- ISGlobal, Barcelona Institute for Global Health, Barcelona, Spain
| | - Vorada Chuenchob
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Niwat Kangwangransan
- Department of Pathobiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Zachary P. Billman
- Department of Laboratory Medicine and Pathology, and Department of Microbiology, University of Washington, Seattle, Washington, USA
| | - Tayla M. Olsen
- Department of Laboratory Medicine and Pathology, and Department of Microbiology, University of Washington, Seattle, Washington, USA
| | - Juan R. González
- ISGlobal, Barcelona Institute for Global Health, Barcelona, Spain
| | - Wanlapa Roobsoong
- MVRU, Mahidol Vivax Research Unit, Mahidol University, Bangkok, Thailand
| | | | - Sean C. Murphy
- Department of Laboratory Medicine and Pathology, and Department of Microbiology, University of Washington, Seattle, Washington, USA
| | - Sebastian A. Mikolajczak
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Eva Borràs
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain,Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Eduard Sabidó
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain,Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Carmen Fernandez-Becerra
- ISGlobal, Barcelona Institute for Global Health, Barcelona, Spain,IGTP, Institute for Health Sciences Trias I Pujol, Barcelona, Spain
| | - Erika L. Flannery
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Stefan H.I. Kappe
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA,Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Hernando A. del Portillo
- ISGlobal, Barcelona Institute for Global Health, Barcelona, Spain,IGTP, Institute for Health Sciences Trias I Pujol, Barcelona, Spain,ICREA, Catalan Institution for Research and Advanced Studies, Barcelona, Spain,For correspondence: Hernando A. del Portillo
| |
Collapse
|
19
|
Ruberto AA, Bourke C, Vantaux A, Maher SP, Jex A, Witkowski B, Snounou G, Mueller I. Single-cell RNA sequencing of Plasmodium vivax sporozoites reveals stage- and species-specific transcriptomic signatures. PLoS Negl Trop Dis 2022; 16:e0010633. [PMID: 35926062 PMCID: PMC9380936 DOI: 10.1371/journal.pntd.0010633] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 08/16/2022] [Accepted: 07/04/2022] [Indexed: 11/24/2022] Open
Abstract
Background Plasmodium vivax sporozoites reside in the salivary glands of a mosquito before infecting a human host and causing malaria. Previous transcriptome-wide studies in populations of these parasite forms were limited in their ability to elucidate cell-to-cell variation, thereby masking cellular states potentially important in understanding malaria transmission outcomes. Methodology/Principal findings In this study, we performed transcription profiling on 9,947 P. vivax sporozoites to assess the extent to which they differ at single-cell resolution. We show that sporozoites residing in the mosquito’s salivary glands exist in distinct developmental states, as defined by their transcriptomic signatures. Additionally, relative to P. falciparum, P. vivax displays overlapping and unique gene usage patterns, highlighting conserved and species-specific gene programs. Notably, distinguishing P. vivax from P. falciparum were a subset of P. vivax sporozoites expressing genes associated with translational regulation and repression. Finally, our comparison of single-cell transcriptomic data from P. vivax sporozoite and erythrocytic forms reveals gene usage patterns unique to sporozoites. Conclusions/Significance In defining the transcriptomic signatures of individual P. vivax sporozoites, our work provides new insights into the factors driving their developmental trajectory and lays the groundwork for a more comprehensive P. vivax cell atlas. Plasmodium vivax is the second most common cause of malaria worldwide. It is particularly challenging for malaria elimination as it forms both active blood-stage infections, as well as asymptomatic liver-stage infections that can persist for extended periods of time. The activation of persister forms in the liver (hypnozoites) are responsible for relapsing infections occurring weeks or months following primary infection via a mosquito bite. How P. vivax persists in the liver remains a major gap in understanding of this organism. It has been hypothesized that there is pre-programming of the infectious sporozoite while it is in the salivary-glands that determines if the cell’s fate once in the liver is to progress towards immediate liver stage development or persist for long-periods as a hypnozoite. The aim of this study was to see if such differences were distinguishable at the transcript level in salivary-gland sporozoites. While we found significant variation amongst sporozoites, we did not find clear evidence that they are transcriptionally pre-programmed as has been suggested. Nevertheless, we highlight several intriguing patterns that appear to be P. vivax specific relative to non-relapsing species that cause malaria prompting further investigation.
Collapse
Affiliation(s)
- Anthony A. Ruberto
- Department of Parasites and Insect Vectors, Institut Pasteur, Paris, France
| | - Caitlin Bourke
- Division of Population Health and Immunity, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Amélie Vantaux
- Malaria Molecular Epidemiology Unit, Institut Pasteur du Cambodge, Phnom Penh, Kingdom of Cambodia
| | - Steven P. Maher
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, United States of America
| | - Aaron Jex
- Division of Population Health and Immunity, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Benoit Witkowski
- Malaria Molecular Epidemiology Unit, Institut Pasteur du Cambodge, Phnom Penh, Kingdom of Cambodia
| | - Georges Snounou
- Commissariat à l’Énergie Atomique et aux Énergies Alternatives-Université Paris Sud 11-INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases (IMVA-HB), Infectious Disease Models and Innovative Therapies (IDMIT) Department, Institut de Biologie François Jacob (IBFJ), Direction de la Recherche Fondamentale (DRF), Fontenay-aux-Roses, France
| | - Ivo Mueller
- Department of Parasites and Insect Vectors, Institut Pasteur, Paris, France
- Division of Population Health and Immunity, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
- * E-mail:
| |
Collapse
|
20
|
Pomun T, Wonginta P, Kubera A. Malaria Box Compounds against Anopheles gambiae (Diptera: Culicidae) Carboxypeptidase B Activity to Block Malaria Transmission. JOURNAL OF MEDICAL ENTOMOLOGY 2022; 59:1355-1362. [PMID: 35522203 DOI: 10.1093/jme/tjac043] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Indexed: 06/14/2023]
Abstract
Carboxypeptidase B (CPB) plays an important role in blood digestion in mosquitos, aiding the release of free amino acids. Anopheles CPB is a target to block malaria transmission because it facilitates Plasmodium invasion of the mosquito midgut. Our study aimed to discover inhibitors of Anopheles CPB to prevent Plasmodium development in the mosquito. The Anopheles gambiae cpb (Agcpb) gene without a signal sequence was cloned into the pET28b expression vector. The recombinant AgCPB protein was expressed in E. coli BL21(DE3) within inclusion bodies after induction with 0.5 mM isopropyl β-D-1-thiogalactopyranoside at 37°C for 4 h. The protein pellet was dissolved in 6 M urea, purified by affinity chromatography, and dialyzed in reaction buffer. The refolded recombinant AgCPB could digest the hippuryl-arginine substrate similarly to that of the commercial porcine pancreas CPB. The 20 top-scoring malaria box compounds from the virtual-screening results were then chosen for an in vitro inhibition assay against AgCPB. Four of the 20 malaria box compounds could inhibit AgCPB activity. The compound MMV007591 was the most potent inhibitor with an IC50 at 0.066 µM. The results indicate that these candidate compounds may be utilized in drug development against mosquito CPB activity to curb malaria transmission.
Collapse
Affiliation(s)
- Tippawan Pomun
- Department of Genetics, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
| | - Phattaradanai Wonginta
- Department of Genetics, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
| | - Anchanee Kubera
- Department of Genetics, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
- Centre for Advanced Studies in Tropical Natural Resources, Kasetsart University, Bangkok 10900, Thailand
| |
Collapse
|
21
|
Abstract
"The Primate Malarias" book has been a uniquely important resource for multiple generations of scientists, since its debut in 1971, and remains pertinent to the present day. Indeed, nonhuman primates (NHPs) have been instrumental for major breakthroughs in basic and pre-clinical research on malaria for over 50 years. Research involving NHPs have provided critical insights and data that have been essential for malaria research on many parasite species, drugs, vaccines, pathogenesis, and transmission, leading to improved clinical care and advancing research goals for malaria control, elimination, and eradication. Whilst most malaria scientists over the decades have been studying Plasmodium falciparum, with NHP infections, in clinical studies with humans, or using in vitro culture or rodent model systems, others have been dedicated to advancing research on Plasmodium vivax, as well as on phylogenetically related simian species, including Plasmodium cynomolgi, Plasmodium coatneyi, and Plasmodium knowlesi. In-depth study of these four phylogenetically related species over the years has spawned the design of NHP longitudinal infection strategies for gathering information about ongoing infections, which can be related to human infections. These Plasmodium-NHP infection model systems are reviewed here, with emphasis on modern systems biological approaches to studying longitudinal infections, pathogenesis, immunity, and vaccines. Recent discoveries capitalizing on NHP longitudinal infections include an advanced understanding of chronic infections, relapses, anaemia, and immune memory. With quickly emerging new technological advances, more in-depth research and mechanistic discoveries can be anticipated on these and additional critical topics, including hypnozoite biology, antigenic variation, gametocyte transmission, bone marrow dysfunction, and loss of uninfected RBCs. New strategies and insights published by the Malaria Host-Pathogen Interaction Center (MaHPIC) are recapped here along with a vision that stresses the importance of educating future experts well trained in utilizing NHP infection model systems for the pursuit of innovative, effective interventions against malaria.
Collapse
Affiliation(s)
- Mary R Galinski
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA.
- Emory Vaccine Center, Emory University, Atlanta, GA, USA.
- Emory National Primate Research Center (Yerkes National Primate Research Center), Emory University, Atlanta, GA, USA.
| |
Collapse
|
22
|
New insights into antimalarial chemopreventive activity of antifolates. Antimicrob Agents Chemother 2021; 66:e0153821. [PMID: 34930029 DOI: 10.1128/aac.01538-21] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Antifolates targeting dihydrofolate reductase (DHFR) are antimalarial compounds that have long been used for malaria treatment and chemoprevention (inhibition of infection from mosquitoes to humans). Despite their extensive applications, the thorough understanding of antifolate activity against hepatic malaria parasites, especially resistant parasites, have yet to be achieved. Using a transgenic P. berghei harboring quadruple mutant dhfr from P. falciparum (Pb::Pfdhfr-4M), we demonstrate that quadruple mutations on Pfdhfr confer complete chemoprevention resistance to pyrimethamine, the previous generation of antifolate, but not to a new class of antifolate designed to overcome the resistance such as P218. Detailed investigation to pin-point stage-specific chemoprevention further demonstrated that it is unnecessary for the drug to be present throughout hepatic development. The drug is most potent against the developmental stages from early hepatic trophozoite to late hepatic trophozoite, but is not effective at inhibiting sporozoite and early hepatic stage development from sporozoite to early trophozoite. Our data shows that P218 also inhibited the late hepatic stage development, from trophozoite to mature schizonts to a lesser extent. With a single dose of 15 mg/kg, P218 prevented infection from up to 25,000 pyrimethamine-resistant sporozoites, a number equal to thousands of infectious mosquito bites. Additionally, the hepatic stage of malaria parasite is much more susceptible to antifolates than the asexual blood stage. This study provides important insights into the activity of antifolates, as a chemopreventive therapeutic which could lead to a more efficient and cost effective treatment regime.
Collapse
|
23
|
Zanghi G, Vaughan AM. Plasmodium vivax pre-erythrocytic stages and the latent hypnozoite. Parasitol Int 2021; 85:102447. [PMID: 34474178 DOI: 10.1016/j.parint.2021.102447] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 08/24/2021] [Accepted: 08/26/2021] [Indexed: 02/02/2023]
Abstract
Plasmodium vivax is the most geographically widespread malaria parasite on the planet. This is largely because after mosquito transmission, P. vivax sporozoites can invade hepatocytes and form latent liver stages known as hypnozoites. These persistent liver stages can activate weeks, months or even years after an infected individual suffers a primary clinical infection. Activation then leads to replication and liver stage schizont maturation that ultimately cause relapse of blood stage infection, disease, and onward transmission. Thus, the latent hypnozoite can lie in wait during times when onward transmission is unlikely due to conditions that do not favor the mosquito. For example, in temperate climates where mosquito prevalence is only seasonal. Furthermore, the elimination of hypnozoites is challenging since the hypnozoite reservoir is currently undetectable and not killed by most antimalarial drugs. Here, we review our current knowledge of the pre-erythrocytic stages of the malaria parasite - the sporozoite and liver stages, including the elusive and enigmatic hypnozoite. We focus on our understanding of sporozoite biology, the novel animal models that are available to study the hypnozoite and hypnozoite activation and the ongoing efforts to understand the biological makeup of the hypnozoite that allow for its persistence in the human host.
Collapse
Affiliation(s)
| | - Ashley M Vaughan
- Seattle Children's Research Institute, Seattle, WA, USA; Department of Pediatrics, University of Washington, Seattle, WA, USA.
| |
Collapse
|
24
|
Schäfer C, Zanghi G, Vaughan AM, Kappe SHI. Plasmodium vivax Latent Liver Stage Infection and Relapse: Biological Insights and New Experimental Tools. Annu Rev Microbiol 2021; 75:87-106. [PMID: 34196569 DOI: 10.1146/annurev-micro-032421-061155] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Plasmodium vivax is the most widespread human malaria parasite, in part because it can form latent liver stages known as hypnozoites after transmission by female anopheline mosquitoes to human hosts. These persistent stages can activate weeks, months, or even years after the primary clinical infection; replicate; and initiate relapses of blood stage infection, which causes disease and recurring transmission. Eliminating hypnozoites is a substantial obstacle for malaria treatment and eradication since the hypnozoite reservoir is undetectable and unaffected by most antimalarial drugs. Importantly, in some parts of the globe where P. vivax malaria is endemic, as many as 90% of P. vivax blood stage infections are thought to be relapses rather than primary infections, rendering the hypnozoite a major driver of P. vivax epidemiology. Here, we review the biology of the hypnozoite and recent discoveries concerning this enigmatic parasite stage. We discuss treatment and prevention challenges, novel animal models to study hypnozoites and relapse, and hypotheses related to hypnozoite formation and activation. Expected final online publication date for the Annual Review of Microbiology, Volume 75 is October 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Carola Schäfer
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington 98109, USA; , , ,
| | - Gigliola Zanghi
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington 98109, USA; , , ,
| | - Ashley M Vaughan
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington 98109, USA; , , , .,Department of Pediatrics, University of Washington, Seattle, Washington 98105, USA
| | - Stefan H I Kappe
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington 98109, USA; , , , .,Department of Pediatrics, University of Washington, Seattle, Washington 98105, USA.,Deparment of Global Health, University of Washington, Seattle, Washington 98195, USA
| |
Collapse
|
25
|
Real E, Howick VM, Dahalan FA, Witmer K, Cudini J, Andradi-Brown C, Blight J, Davidson MS, Dogga SK, Reid AJ, Baum J, Lawniczak MKN. A single-cell atlas of Plasmodium falciparum transmission through the mosquito. Nat Commun 2021; 12:3196. [PMID: 34045457 PMCID: PMC8159942 DOI: 10.1038/s41467-021-23434-z] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 04/28/2021] [Indexed: 01/29/2023] Open
Abstract
Malaria parasites have a complex life cycle featuring diverse developmental strategies, each uniquely adapted to navigate specific host environments. Here we use single-cell transcriptomics to illuminate gene usage across the transmission cycle of the most virulent agent of human malaria - Plasmodium falciparum. We reveal developmental trajectories associated with the colonization of the mosquito midgut and salivary glands and elucidate the transcriptional signatures of each transmissible stage. Additionally, we identify both conserved and non-conserved gene usage between human and rodent parasites, which point to both essential mechanisms in malaria transmission and species-specific adaptations potentially linked to host tropism. Together, the data presented here, which are made freely available via an interactive website, provide a fine-grained atlas that enables intensive investigation of the P. falciparum transcriptional journey. As well as providing insights into gene function across the transmission cycle, the atlas opens the door for identification of drug and vaccine targets to stop malaria transmission and thereby prevent disease.
Collapse
Affiliation(s)
- Eliana Real
- Department of Life Sciences, Imperial College London, London, UK
| | - Virginia M Howick
- Parasites and Microbes Programme, Wellcome Sanger Institute, Hinxton, UK
- Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Wellcome Centre for Integrative Parasitology, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Farah A Dahalan
- Department of Life Sciences, Imperial College London, London, UK
| | - Kathrin Witmer
- Department of Life Sciences, Imperial College London, London, UK
- Parasites and Microbes Programme, Wellcome Sanger Institute, Hinxton, UK
| | - Juliana Cudini
- Parasites and Microbes Programme, Wellcome Sanger Institute, Hinxton, UK
| | - Clare Andradi-Brown
- Department of Life Sciences, Imperial College London, London, UK
- Department of Infectious Disease, Imperial College London, London, UK
| | - Joshua Blight
- Department of Life Sciences, Imperial College London, London, UK
| | - Mira S Davidson
- Department of Life Sciences, Imperial College London, London, UK
| | - Sunil Kumar Dogga
- Parasites and Microbes Programme, Wellcome Sanger Institute, Hinxton, UK
| | - Adam J Reid
- Parasites and Microbes Programme, Wellcome Sanger Institute, Hinxton, UK
| | - Jake Baum
- Department of Life Sciences, Imperial College London, London, UK.
| | - Mara K N Lawniczak
- Parasites and Microbes Programme, Wellcome Sanger Institute, Hinxton, UK.
| |
Collapse
|
26
|
Bourgard C, Lopes SCP, Lacerda MVG, Albrecht L, Costa FTM. A suitable RNA preparation methodology for whole transcriptome shotgun sequencing harvested from Plasmodium vivax-infected patients. Sci Rep 2021; 11:5089. [PMID: 33658571 PMCID: PMC7930272 DOI: 10.1038/s41598-021-84607-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 01/06/2021] [Indexed: 12/03/2022] Open
Abstract
Plasmodium vivax is a world-threatening human malaria parasite, whose biology remains elusive. The unavailability of in vitro culture, and the difficulties in getting a high number of pure parasites makes RNA isolation in quantity and quality a challenge. Here, a methodological outline for RNA-seq from P. vivax isolates with low parasitemia is presented, combining parasite maturation and enrichment with efficient RNA extraction, yielding ~ 100 pg.µL−1 of RNA, suitable for SMART-Seq Ultra-Low Input RNA library and Illumina sequencing. Unbiased coding transcriptome of ~ 4 M reads was achieved for four patient isolates with ~ 51% of transcripts mapped to the P. vivax P01 reference genome, presenting heterogeneous profiles of expression among individual isolates. Amongst the most transcribed genes in all isolates, a parasite-staged mixed repertoire of conserved parasite metabolic, membrane and exported proteins was observed. Still, a quarter of transcribed genes remain functionally uncharacterized. In parallel, a P. falciparum Brazilian isolate was also analyzed and 57% of its transcripts mapped against IT genome. Comparison of transcriptomes of the two species revealed a common trophozoite-staged expression profile, with several homologous genes being expressed. Collectively, these results will positively impact vivax research improving knowledge of P. vivax biology.
Collapse
Affiliation(s)
- Catarina Bourgard
- Laboratory of Tropical Diseases, Prof. Dr. Luiz Jacintho da Silva, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas-UNICAMP, Campinas, SP, Brazil
| | - Stefanie C P Lopes
- Instituto Leônidas & Maria Deane, Fundação Oswaldo Cruz-Fiocruz, Manaus, AM, Brazil.,Fundação de Medicina Tropical Dr. Heitor Vieira Dourado-FMT-HVD, Gerência de Malária, Manaus, AM, Brazil
| | - Marcus V G Lacerda
- Instituto Leônidas & Maria Deane, Fundação Oswaldo Cruz-Fiocruz, Manaus, AM, Brazil.,Fundação de Medicina Tropical Dr. Heitor Vieira Dourado-FMT-HVD, Gerência de Malária, Manaus, AM, Brazil
| | - Letusa Albrecht
- Laboratory of Tropical Diseases, Prof. Dr. Luiz Jacintho da Silva, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas-UNICAMP, Campinas, SP, Brazil. .,Instituto Carlos Chagas, Fundação Oswaldo Cruz-Fiocruz, Curitiba, PR, Brazil.
| | - Fabio T M Costa
- Laboratory of Tropical Diseases, Prof. Dr. Luiz Jacintho da Silva, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas-UNICAMP, Campinas, SP, Brazil.
| |
Collapse
|
27
|
Ruberto AA, Bourke C, Merienne N, Obadia T, Amino R, Mueller I. Single-cell RNA sequencing reveals developmental heterogeneity among Plasmodium berghei sporozoites. Sci Rep 2021; 11:4127. [PMID: 33619283 PMCID: PMC7900125 DOI: 10.1038/s41598-021-82914-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 01/27/2021] [Indexed: 12/17/2022] Open
Abstract
In the malaria-causing parasite's life cycle, Plasmodium sporozoites must travel from the midgut of a mosquito to the salivary glands before they can infect a mammalian host. However, only a fraction of sporozoites complete the journey. Since salivary gland invasion is required for transmission of sporozoites, insights at the molecular level can contribute to strategies for malaria prevention. Recent advances in single-cell RNA sequencing provide an opportunity to assess sporozoite heterogeneity at a resolution unattainable by bulk RNA sequencing methods. In this study, we use a droplet-based single-cell RNA sequencing workflow to analyze the transcriptomes of over 8000 Plasmodium berghei sporozoites derived from the midguts and salivary glands of Anopheles stephensi mosquitoes. The detection of known marker genes confirms the successful capture and sequencing of samples composed of a mixed population of sporozoites. Using data integration, clustering, and trajectory analyses, we reveal differences in gene expression profiles of individual sporozoites, and identify both annotated and unannotated markers associated with sporozoite development. Our work highlights the utility of a high-throughput workflow for the transcriptomic profiling of Plasmodium sporozoites, and provides new insights into gene usage during the parasite's development in the mosquito.
Collapse
Affiliation(s)
- Anthony A Ruberto
- Department of Parasites and Insect Vectors, Institut Pasteur, Paris, France.
| | - Caitlin Bourke
- Division of Population Health and Immunity, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Nicolas Merienne
- Department of Parasites and Insect Vectors, Institut Pasteur, Paris, France
| | - Thomas Obadia
- Department of Parasites and Insect Vectors, Institut Pasteur, Paris, France
- Hub de Bioinformatique et Biostatistique - Département Biologie Computationnelle, Institut Pasteur, 75015, Paris, France
| | - Rogerio Amino
- Department of Parasites and Insect Vectors, Institut Pasteur, Paris, France
| | - Ivo Mueller
- Department of Parasites and Insect Vectors, Institut Pasteur, Paris, France.
- Division of Population Health and Immunity, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.
- Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
28
|
Kumari S, Chauhan C, Tevatiya S, Singla D, De TD, Sharma P, Thomas T, Rani J, Savargaonkar D, Pandey KC, Pande V, Dixit R. Genetic changes of Plasmodium vivax tempers host tissue-specific responses in Anopheles stephensi. CURRENT RESEARCH IN IMMUNOLOGY 2021; 2:12-22. [PMID: 35492403 PMCID: PMC9040150 DOI: 10.1016/j.crimmu.2021.02.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 01/13/2021] [Accepted: 02/14/2021] [Indexed: 02/02/2023] Open
Abstract
Recently, we showed how an early restriction of gut flora proliferation by Plasmodium vivax favors immune-suppression and Plasmodium survival in the gut lumen (Sharma et al., 2020). Here, we asked post gut invasion how P. vivax interacts with individual tissues such as the midgut, hemocyte, and salivary glands, and manages its survival in the mosquito host. Our data from tissue-specific comparative RNA-Seq analysis and extensive temporal/spatial expression profiling of selected mosquito transcripts in the uninfected and P. vivax infected mosquito's tissues indicated that (i) a transient suppression of gut metabolic machinery by early oocysts; (ii) enriched expression of nutritional responsive proteins and immune proteins against late oocysts, together may ensure optimal parasite development and gut homeostasis restoration; (iii) pre-immune activation of hemocyte by early gut-oocysts infection via REL induction (p < 0.003); and altered expression of hemocyte-encoded immune proteins may cause rapid removal of free circulating sporozoites from hemolymph; (iv) while a strong suppression of salivary metabolic activities, and elevated expression of salivary specific secretory, as well as immune proteins together, may favor the long-term storage and survival of invaded sporozoites. Finally, our RNA-Seq-based discovery of 4449 transcripts of Plasmodium vivax origin, and their developmental stage-specific expression modulation in the corresponding infected mosquito tissues, predicts a possible mechanism of mosquito responses evasion by P. vivax. Conclusively, our system-wide RNA-Seq analysis provides the first genetic evidence of direct mosquito-Plasmodium interaction and establishes a functional correlation.
Collapse
Affiliation(s)
- Seena Kumari
- Laboratory of Host-Parasite Interaction Studies, ICMR-National Institute of Malaria Research, Dwarka, New Delhi, 110077, India
| | - Charu Chauhan
- Laboratory of Host-Parasite Interaction Studies, ICMR-National Institute of Malaria Research, Dwarka, New Delhi, 110077, India
| | - Sanjay Tevatiya
- Laboratory of Host-Parasite Interaction Studies, ICMR-National Institute of Malaria Research, Dwarka, New Delhi, 110077, India
| | - Deepak Singla
- Laboratory of Host-Parasite Interaction Studies, ICMR-National Institute of Malaria Research, Dwarka, New Delhi, 110077, India
- School of Agricultural Biotechnology, Punjab Agricultural University, Ludhiana, Punjab, India
| | - Tanwee Das De
- Laboratory of Host-Parasite Interaction Studies, ICMR-National Institute of Malaria Research, Dwarka, New Delhi, 110077, India
| | - Punita Sharma
- Laboratory of Host-Parasite Interaction Studies, ICMR-National Institute of Malaria Research, Dwarka, New Delhi, 110077, India
| | - Tina Thomas
- Laboratory of Host-Parasite Interaction Studies, ICMR-National Institute of Malaria Research, Dwarka, New Delhi, 110077, India
| | - Jyoti Rani
- Laboratory of Host-Parasite Interaction Studies, ICMR-National Institute of Malaria Research, Dwarka, New Delhi, 110077, India
- Bio and Nanotechnology Department, Guru Jambheshwar University of Science and Technology, Haryana, India
| | - Deepali Savargaonkar
- Laboratory of Host-Parasite Interaction Studies, ICMR-National Institute of Malaria Research, Dwarka, New Delhi, 110077, India
| | - Kailash C. Pandey
- Laboratory of Host-Parasite Interaction Studies, ICMR-National Institute of Malaria Research, Dwarka, New Delhi, 110077, India
| | - Veena Pande
- Department of Biotechnology, Kumaun University, Nainital, Uttarakhand, India
| | - Rajnikant Dixit
- Laboratory of Host-Parasite Interaction Studies, ICMR-National Institute of Malaria Research, Dwarka, New Delhi, 110077, India
| |
Collapse
|
29
|
Briquet S, Marinach C, Silvie O, Vaquero C. Preparing for Transmission: Gene Regulation in Plasmodium Sporozoites. Front Cell Infect Microbiol 2021; 10:618430. [PMID: 33585284 PMCID: PMC7878544 DOI: 10.3389/fcimb.2020.618430] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/16/2020] [Indexed: 11/13/2022] Open
Abstract
Plasmodium sporozoites are transmitted to mammals by anopheline mosquitoes and first infect the liver, where they transform into replicative exoerythrocytic forms, which subsequently release thousands of merozoites that invade erythrocytes and initiate the malaria disease. In some species, sporozoites can transform into dormant hypnozoites in the liver, which cause malaria relapses upon reactivation. Transmission from the insect vector to a mammalian host is a critical step of the parasite life cycle, and requires tightly regulated gene expression. Sporozoites are formed inside oocysts in the mosquito midgut and become fully infectious after colonization of the insect salivary glands, where they remain quiescent until transmission. Parasite maturation into infectious sporozoites is associated with reprogramming of the sporozoite transcriptome and proteome, which depends on multiple layers of transcriptional and post-transcriptional regulatory mechanisms. An emerging scheme is that gene expression in Plasmodium sporozoites is controlled by alternating waves of transcription activity and translational repression, which shape the parasite RNA and protein repertoires for successful transition from the mosquito vector to the mammalian host.
Collapse
Affiliation(s)
- Sylvie Briquet
- Centre d'Immunologie et des Maladies Infectieuses, INSERM, CNRS, Sorbonne Université, Paris, France
| | - Carine Marinach
- Centre d'Immunologie et des Maladies Infectieuses, INSERM, CNRS, Sorbonne Université, Paris, France
| | - Olivier Silvie
- Centre d'Immunologie et des Maladies Infectieuses, INSERM, CNRS, Sorbonne Université, Paris, France
| | - Catherine Vaquero
- Centre d'Immunologie et des Maladies Infectieuses, INSERM, CNRS, Sorbonne Université, Paris, France
| |
Collapse
|
30
|
Kumari S, Chauhan C, Tevatiya S, Singla D, De TD, Sharma P, Thomas T, Rani J, Savargaonkar D, Pandey KC, Pande V, Dixit R. Genetic changes of Plasmodium vivax tempers host tissue-specific responses in Anopheles stephensi. CURRENT RESEARCH IN IMMUNOLOGY 2021. [DOI: https:/doi.org/10.1016/j.crimmu.2021.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
|
31
|
Kumari S, Chauhan C, Tevatiya S, Singla D, De TD, Sharma P, Thomas T, Rani J, Savargaonkar D, Pandey KC, Pande V, Dixit R. Genetic changes of Plasmodium vivax tempers host tissue-specific responses in Anopheles stephensi. CURRENT RESEARCH IN IMMUNOLOGY 2021. [DOI: https://doi.org/10.1016/j.crimmu.2021.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
|
32
|
Boonkaew T, Mongkol W, Prasert S, Paochan P, Yoneda S, Nguitragool W, Kumpitak C, Sattabongkot J, Kubera A. Transcriptome analysis of Anopheles dirus and Plasmodium vivax at ookinete and oocyst stages. Acta Trop 2020; 207:105502. [PMID: 32320680 DOI: 10.1016/j.actatropica.2020.105502] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 04/15/2020] [Accepted: 04/15/2020] [Indexed: 02/07/2023]
Abstract
Malaria is transmitted by Plasmodium parasites through the bite of female Anopheles mosquitoes. One of the most important mosquito vectors in the Greater Mekong Subregion is Anopheles dirus. This study reports RNA sequencing (RNA-Seq) transcriptome analysis of An. dirus at 18 hours and 7 days after a P. vivax-infected blood meal, which represent infection at the ookinete and oocyst parasite developmental stages, respectively. Following infection, 582 An. dirus transcripts were modulated. The 2,408 P. vivax transcripts could be classified into ookinete-specific, two-stage, and oocyst-specific groups. Results were validated by quantitative reverse transcription polymerase chain reaction. Gene ontology analysis of the vector and parasite revealed several biological pathways for both, providing a better understanding of Anopheles-Plasmodium interactions at the ookinete and oocyst stages.
Collapse
Affiliation(s)
- Tippawan Boonkaew
- Department of Genetics, Faculty of Science, Kasetsart University, Bangkok, 10900, Thailand
| | - Watcharakorn Mongkol
- Department of Genetics, Faculty of Science, Kasetsart University, Bangkok, 10900, Thailand
| | - Sureerat Prasert
- Department of Genetics, Faculty of Science, Kasetsart University, Bangkok, 10900, Thailand
| | - Pattaweeya Paochan
- Department of Genetics, Faculty of Science, Kasetsart University, Bangkok, 10900, Thailand
| | - Saki Yoneda
- Department of Biotechnology, Graduate school of Engineering, Osaka University, Japan
| | - Wang Nguitragool
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
| | - Chalermpon Kumpitak
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
| | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
| | - Anchanee Kubera
- Department of Genetics, Faculty of Science, Kasetsart University, Bangkok, 10900, Thailand; Centre for Advanced Studies in Tropical Natural Resources, Kasetsart University, Bangkok, 10900, Thailand.
| |
Collapse
|
33
|
Medeiros CMP, Moreira EUM, Pires CV, Torres LM, Guimarães LFF, Alves JRS, Lima BAS, Fontes CJF, Costa HL, Brito CFA, Sousa TN, Ntumngia FB, Adams JH, Kano FS, Carvalho LH. Dynamics of IgM and IgG responses to the next generation of engineered Duffy binding protein II immunogen: Strain-specific and strain-transcending immune responses over a nine-year period. PLoS One 2020; 15:e0232786. [PMID: 32379804 PMCID: PMC7205269 DOI: 10.1371/journal.pone.0232786] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 04/21/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND A low proportion of P. vivax-exposed individuals acquire protective strain-transcending neutralizing IgG antibodies that are able to block the interaction between the Duffy binding protein II (DBPII) and its erythrocyte-specific invasion receptor. In a recent study, a novel surface-engineered DBPII-based vaccine termed DEKnull-2, whose antibody response target conserved DBPII epitopes, was able to induce broadly binding-inhibitory IgG antibodies (BIAbs) that inhibit P. vivax reticulocyte invasion. Toward the development of DEKnull-2 as an effective P. vivax blood-stage vaccine, we investigate the relationship between naturally acquired DBPII-specific IgM response and the profile of IgG antibodies/BIAbs activity over time. METHODOLOGY/PRINCIPAL FINDINGS A nine-year follow-up study was carried-out among long-term P. vivax-exposed Amazonian individuals and included six cross-sectional surveys at periods of high and low malaria transmission. DBPII immune responses associated with either strain-specific (Sal1, natural DBPII variant circulating in the study area) or conserved epitopes (DEKnull-2) were monitored by conventional serology (ELISA-detected IgM and IgG antibodies), with IgG BIAbs activity evaluated by functional assays (in vitro inhibition of DBPII-erythrocyte binding). The results showed a tendency of IgM antibodies toward Sal1-specific response; the profile of Sal1 over DEKnull-2 was not associated with acute malaria and sustained throughout the observation period. The low malaria incidence in two consecutive years allowed us to demonstrate that variant-specific IgG (but not IgM) antibodies waned over time, which resulted in IgG skewed to the DEKnull-2 response. A persistent DBPII-specific IgM response was not associated with the presence (or absence) of broadly neutralizing IgG antibody response. CONCLUSIONS/SIGNIFICANCE The current study demonstrates that long-term exposure to low and unstable levels of P. vivax transmission led to a sustained DBPII-specific IgM response against variant-specific epitopes, while sustained IgG responses are skewed to conserved epitopes. Further studies should investigate on the role of a stable and persistent IgM antibody response in the immune response mediated by DBPII.
Collapse
Affiliation(s)
- Camila M. P. Medeiros
- Centro de Pesquisas René Rachou/FIOCRUZ Minas, Belo Horizonte, MG, Brazil
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Camilla V. Pires
- Centro de Pesquisas René Rachou/FIOCRUZ Minas, Belo Horizonte, MG, Brazil
| | - Letícia M. Torres
- Centro de Pesquisas René Rachou/FIOCRUZ Minas, Belo Horizonte, MG, Brazil
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | | | | | - Bárbara A. S. Lima
- Centro de Pesquisas René Rachou/FIOCRUZ Minas, Belo Horizonte, MG, Brazil
| | - Cor J. F. Fontes
- Hospital Júlio Muller, Universidade Federal de Mato Grosso, Cuiabá, Mato Grosso, Brazil
| | - Helena L. Costa
- Centro de Pesquisas René Rachou/FIOCRUZ Minas, Belo Horizonte, MG, Brazil
| | | | - Tais N. Sousa
- Centro de Pesquisas René Rachou/FIOCRUZ Minas, Belo Horizonte, MG, Brazil
| | - Francis B. Ntumngia
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, Florida, United States of America
| | - John H. Adams
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, Florida, United States of America
| | - Flora S. Kano
- Centro de Pesquisas René Rachou/FIOCRUZ Minas, Belo Horizonte, MG, Brazil
- * E-mail: (LHC); (FSK)
| | - Luzia H. Carvalho
- Centro de Pesquisas René Rachou/FIOCRUZ Minas, Belo Horizonte, MG, Brazil
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
- * E-mail: (LHC); (FSK)
| |
Collapse
|
34
|
Posfai D, Maher SP, Roesch C, Vantaux A, Sylvester K, Péneau J, Popovici J, Kyle DE, Witkowski B, Derbyshire ER. Plasmodium vivax Liver and Blood Stages Recruit the Druggable Host Membrane Channel Aquaporin-3. Cell Chem Biol 2020; 27:719-727.e5. [PMID: 32330444 PMCID: PMC7303948 DOI: 10.1016/j.chembiol.2020.03.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 02/07/2020] [Accepted: 03/09/2020] [Indexed: 12/17/2022]
Abstract
Plasmodium vivax infects hepatocytes to form schizonts that cause blood infection, or dormant hypnozoites that can persist for months in the liver before leading to relapsing blood infections. The molecular processes that drive P. vivax schizont and hypnozoite survival remain largely unknown, but they likely involve a rich network of host-pathogen interactions, including those occurring at the host-parasite interface, the parasitophorous vacuole membrane (PVM). Using a recently developed P. vivax liver-stage model system we demonstrate that host aquaporin-3 (AQP3) localizes to the PVM of schizonts and hypnozoites within 5 days after invasion. This recruitment is also observed in P. vivax-infected reticulocytes. Chemical treatment with the AQP3 inhibitor auphen reduces P. vivax liver hypnozoite and schizont burden, and inhibits P. vivax asexual blood-stage growth. These findings reveal a role for AQP3 in P. vivax liver and blood stages and suggest that the protein may be targeted for therapeutic treatment. Host aquaporin-3 (AQP3) is recruited to P. vivax hypnozoites and schizonts The AQP3 inhibitor auphen inhibits P. vivax hypnozoites and schizonts Host AQP3 is recruited to P. vivax-infected erythrocytes derived from patient samples Auphen inhibits blood stages of clinical P. vivax isolates
Collapse
Affiliation(s)
- Dora Posfai
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, 213 Research Drive, Durham, NC 27710, USA
| | - Steven P Maher
- Center for Tropical and Emerging Global Diseases, University of Georgia, 500 D.W. Brooks Dr, ste 370, Athens, GE 30602, USA
| | - Camille Roesch
- Malaria Molecular Epidemiology Unit, Pasteur Institute in Cambodia, Phnom Penh 12201, Cambodia
| | - Amélie Vantaux
- Malaria Molecular Epidemiology Unit, Pasteur Institute in Cambodia, Phnom Penh 12201, Cambodia
| | - Kayla Sylvester
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, 213 Research Drive, Durham, NC 27710, USA
| | - Julie Péneau
- Malaria Molecular Epidemiology Unit, Pasteur Institute in Cambodia, Phnom Penh 12201, Cambodia
| | - Jean Popovici
- Malaria Molecular Epidemiology Unit, Pasteur Institute in Cambodia, Phnom Penh 12201, Cambodia
| | - Dennis E Kyle
- Center for Tropical and Emerging Global Diseases, University of Georgia, 500 D.W. Brooks Dr, ste 370, Athens, GE 30602, USA
| | - Benoît Witkowski
- Malaria Molecular Epidemiology Unit, Pasteur Institute in Cambodia, Phnom Penh 12201, Cambodia.
| | - Emily R Derbyshire
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, 213 Research Drive, Durham, NC 27710, USA; Chemistry Department, Duke University, 124 Science Drive, Durham, NC 27708, USA.
| |
Collapse
|
35
|
Ruiz JL, Gómez-Díaz E. The second life of Plasmodium in the mosquito host: gene regulation on the move. Brief Funct Genomics 2020; 18:313-357. [PMID: 31058281 DOI: 10.1093/bfgp/elz007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 03/08/2019] [Accepted: 03/26/2019] [Indexed: 01/08/2023] Open
Abstract
Malaria parasites face dynamically changing environments and strong selective constraints within human and mosquito hosts. To survive such hostile and shifting conditions, Plasmodium switches transcriptional programs during development and has evolved mechanisms to adjust its phenotype through heterogeneous patterns of gene expression. In vitro studies on culture-adapted isolates have served to set the link between chromatin structure and functional gene expression. Yet, experimental evidence is limited to certain stages of the parasite in the vertebrate, i.e. blood, while the precise mechanisms underlying the dynamic regulatory landscapes during development and in the adaptation to within-host conditions remain poorly understood. In this review, we discuss available data on transcriptional and epigenetic regulation in Plasmodium mosquito stages in the context of sporogonic development and phenotypic variation, including both bet-hedging and environmentally triggered direct transcriptional responses. With this, we advocate the mosquito offers an in vivo biological model to investigate the regulatory networks, transcription factors and chromatin-modifying enzymes and their modes of interaction with regulatory sequences, which might be responsible for the plasticity of the Plasmodium genome that dictates stage- and cell type-specific blueprints of gene expression.
Collapse
Affiliation(s)
- José L Ruiz
- Instituto de Parasitología y Biomedicina López-Neyra (IPBLN), Consejo Superior de Investigaciones Científicas, Granada, Spain
| | - Elena Gómez-Díaz
- Instituto de Parasitología y Biomedicina López-Neyra (IPBLN), Consejo Superior de Investigaciones Científicas, Granada, Spain
| |
Collapse
|
36
|
Favuzza P, de Lera Ruiz M, Thompson JK, Triglia T, Ngo A, Steel RWJ, Vavrek M, Christensen J, Healer J, Boyce C, Guo Z, Hu M, Khan T, Murgolo N, Zhao L, Penington JS, Reaksudsan K, Jarman K, Dietrich MH, Richardson L, Guo KY, Lopaticki S, Tham WH, Rottmann M, Papenfuss T, Robbins JA, Boddey JA, Sleebs BE, Sabroux HJ, McCauley JA, Olsen DB, Cowman AF. Dual Plasmepsin-Targeting Antimalarial Agents Disrupt Multiple Stages of the Malaria Parasite Life Cycle. Cell Host Microbe 2020; 27:642-658.e12. [PMID: 32109369 PMCID: PMC7146544 DOI: 10.1016/j.chom.2020.02.005] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 12/22/2019] [Accepted: 02/11/2020] [Indexed: 01/07/2023]
Abstract
Artemisin combination therapy (ACT) is the main treatment option for malaria, which is caused by the intracellular parasite Plasmodium. However, increased resistance to ACT highlights the importance of finding new drugs. Recently, the aspartic proteases Plasmepsin IX and X (PMIX and PMX) were identified as promising drug targets. In this study, we describe dual inhibitors of PMIX and PMX, including WM382, that block multiple stages of the Plasmodium life cycle. We demonstrate that PMX is a master modulator of merozoite invasion and direct maturation of proteins required for invasion, parasite development, and egress. Oral administration of WM382 cured mice of P. berghei and prevented blood infection from the liver. In addition, WM382 was efficacious against P. falciparum asexual infection in humanized mice and prevented transmission to mosquitoes. Selection of resistant P. falciparum in vitro was not achievable. Together, these show that dual PMIX and PMX inhibitors are promising candidates for malaria treatment and prevention.
Collapse
Affiliation(s)
- Paola Favuzza
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; University of Melbourne, Melbourne, VIC 3010, Australia
| | | | - Jennifer K Thompson
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Tony Triglia
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Anna Ngo
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Ryan W J Steel
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; University of Melbourne, Melbourne, VIC 3010, Australia
| | - Marissa Vavrek
- Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA 19486, USA
| | - Janni Christensen
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; University of Melbourne, Melbourne, VIC 3010, Australia
| | - Julie Healer
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; University of Melbourne, Melbourne, VIC 3010, Australia
| | | | - Zhuyan Guo
- Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA 19486, USA
| | - Mengwei Hu
- Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA 19486, USA
| | - Tanweer Khan
- Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA 19486, USA
| | - Nicholas Murgolo
- Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA 19486, USA
| | - Lianyun Zhao
- Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA 19486, USA
| | | | - Kitsanapong Reaksudsan
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; University of Melbourne, Melbourne, VIC 3010, Australia
| | - Kate Jarman
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Melanie H Dietrich
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; University of Melbourne, Melbourne, VIC 3010, Australia
| | - Lachlan Richardson
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; University of Melbourne, Melbourne, VIC 3010, Australia
| | - Kai-Yuan Guo
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; University of Melbourne, Melbourne, VIC 3010, Australia
| | - Sash Lopaticki
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Wai-Hong Tham
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; University of Melbourne, Melbourne, VIC 3010, Australia
| | | | - Tony Papenfuss
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; University of Melbourne, Melbourne, VIC 3010, Australia
| | | | - Justin A Boddey
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; University of Melbourne, Melbourne, VIC 3010, Australia
| | - Brad E Sleebs
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; University of Melbourne, Melbourne, VIC 3010, Australia
| | - Hélène Jousset Sabroux
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; University of Melbourne, Melbourne, VIC 3010, Australia
| | - John A McCauley
- Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA 19486, USA
| | - David B Olsen
- Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA 19486, USA.
| | - Alan F Cowman
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; University of Melbourne, Melbourne, VIC 3010, Australia.
| |
Collapse
|
37
|
Maher SP, Conway AJ, Roth A, Adapa SR, Cualing P, Andolina C, Hsiao J, Turgeon J, Chaumeau V, Johnson M, Palmiotti C, Singh N, Barnes SJ, Patel R, Van Grod V, Carter R, Sun HCS, Sattabongkot J, Campo B, Nosten F, Saadi WM, Adams JH, Jiang RHY, Kyle DE. An adaptable soft-mold embossing process for fabricating optically-accessible, microfeature-based culture systems and application toward liver stage antimalarial compound testing. LAB ON A CHIP 2020; 20:1124-1139. [PMID: 32055808 DOI: 10.1039/c9lc00921c] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Advanced cell culture methods for modeling organ-level structure have been demonstrated to replicate in vivo conditions more accurately than traditional in vitro cell culture. Given that the liver is particularly important to human health, several advanced culture methods have been developed to experiment with liver disease states, including infection with Plasmodium parasites, the causative agent of malaria. These models have demonstrated that intrahepatic parasites require functionally stable hepatocytes to thrive and robust characterization of the parasite populations' response to investigational therapies is dependent on high-content and high-resolution imaging (HC/RI). We previously reported abiotic confinement extends the functional longevity of primary hepatocytes in a microfluidic platform and set out to instill confinement in a microtiter plate platform while maintaining optical accessibility for HC/RI; with an end-goal of producing an improved P. vivax liver stage culture model. We developed a novel fabrication process in which a PDMS soft mold embosses hepatocyte-confining microfeatures into polystyrene, resulting in microfeature-based hepatocyte confinement (μHEP) slides and plates. Our process was optimized to form both microfeatures and culture wells in a single embossing step, resulting in a 100 μm-thick bottom ideal for HC/RI, and was found inexpensively amendable to microfeature design changes. Microfeatures improved intrahepatic parasite infection rates and μHEP systems were used to reconfirm the activity of reference antimalarials in phenotypic dose-response assays. RNAseq of hepatocytes in μHEP systems demonstrated microfeatures sustain hepatic differentiation and function, suggesting broader utility for preclinical hepatic assays; while our tailorable embossing process could be repurposed for developing additional organ models.
Collapse
Affiliation(s)
- Steven P Maher
- Center for Global Health and Infectious Diseases Research, Department of Global Health, College of Public Health, University of South Florida, Tampa, Florida, USA. and Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, USA
| | - Amy J Conway
- Center for Global Health and Infectious Diseases Research, Department of Global Health, College of Public Health, University of South Florida, Tampa, Florida, USA.
| | - Alison Roth
- Center for Global Health and Infectious Diseases Research, Department of Global Health, College of Public Health, University of South Florida, Tampa, Florida, USA.
| | - Swamy R Adapa
- Center for Global Health and Infectious Diseases Research, Department of Global Health, College of Public Health, University of South Florida, Tampa, Florida, USA.
| | - Phillip Cualing
- Center for Global Health and Infectious Diseases Research, Department of Global Health, College of Public Health, University of South Florida, Tampa, Florida, USA.
| | - Chiara Andolina
- Shoklo Malaria Research Unit, Mahidol Oxford Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand & Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - James Hsiao
- Charles Stark Draper Laboratory, Cambridge, MA, USA
| | - Jessica Turgeon
- Center for Global Health and Infectious Diseases Research, Department of Global Health, College of Public Health, University of South Florida, Tampa, Florida, USA.
| | - Victor Chaumeau
- Shoklo Malaria Research Unit, Mahidol Oxford Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand & Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Myles Johnson
- Center for Global Health and Infectious Diseases Research, Department of Global Health, College of Public Health, University of South Florida, Tampa, Florida, USA.
| | | | - Naresh Singh
- Center for Global Health and Infectious Diseases Research, Department of Global Health, College of Public Health, University of South Florida, Tampa, Florida, USA.
| | - Samantha J Barnes
- Center for Global Health and Infectious Diseases Research, Department of Global Health, College of Public Health, University of South Florida, Tampa, Florida, USA.
| | - Raahil Patel
- Center for Global Health and Infectious Diseases Research, Department of Global Health, College of Public Health, University of South Florida, Tampa, Florida, USA.
| | | | - Robert Carter
- Center for Global Health and Infectious Diseases Research, Department of Global Health, College of Public Health, University of South Florida, Tampa, Florida, USA.
| | | | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Brice Campo
- Medicines for Malaria Venture, Geneva, Switzerland
| | - François Nosten
- Shoklo Malaria Research Unit, Mahidol Oxford Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand & Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | - John H Adams
- Center for Global Health and Infectious Diseases Research, Department of Global Health, College of Public Health, University of South Florida, Tampa, Florida, USA.
| | - Rays H Y Jiang
- Center for Global Health and Infectious Diseases Research, Department of Global Health, College of Public Health, University of South Florida, Tampa, Florida, USA.
| | - Dennis E Kyle
- Center for Global Health and Infectious Diseases Research, Department of Global Health, College of Public Health, University of South Florida, Tampa, Florida, USA. and Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, USA
| |
Collapse
|
38
|
Abstract
The LS of Plasmodium infection is an asymptomatic yet necessary stage for producing blood-infective parasites, the causative agents of malaria. Blocking the liver stage of the life cycle can prevent clinical malaria, but relatively less is known about the parasite’s biology at this stage. Using the rodent model P. berghei, we investigated whole-transcriptome changes occurring as early as 2 hpi of hepatocytes. The transcriptional profiles of early time points (2, 4, 12, and 18 hpi) have not been accessible before due to the technical challenges associated with liver-stage infections. Our data now provide insights into these early parasite fluxes that may facilitate establishment of infection, transformation, and replication in the liver. The apicomplexan parasites Plasmodium spp. are the causative agents of malaria, a disease that poses a significant global health burden. Plasmodium spp. initiate infection of the human host by transforming and replicating within hepatocytes. This liver stage (LS) is poorly understood compared to other Plasmodium life stages, which has hindered our ability to target these parasites for disease prevention. We conducted an extensive transcriptome sequencing (RNA-Seq) analysis throughout the Plasmodium berghei LS, covering as early as 2 h postinfection (hpi) and extending to 48 hpi. Our data revealed that hundreds of genes are differentially expressed at 2 hpi and that multiple genes shown to be important for later infection are upregulated as early as 12 hpi. Using hierarchical clustering along with coexpression analysis, we identified clusters functionally enriched for important liver-stage processes such as interactions with the host cell and redox homeostasis. Furthermore, some of these clusters were highly correlated to the expression of ApiAP2 transcription factors, while showing enrichment of mostly uncharacterized DNA binding motifs. This finding indicates potential LS targets for these transcription factors, while also hinting at alternative uncharacterized DNA binding motifs and transcription factors during this stage. Our work presents a window into the previously undescribed transcriptome of Plasmodium upon host hepatocyte infection to enable a comprehensive view of the parasite’s LS. These findings also provide a blueprint for future studies that extend hypotheses concerning LS gene function in P. berghei to human-infective Plasmodium parasites.
Collapse
|
39
|
Vivax Sporozoite Consortium, Muller I, Jex AR, Kappe SHI, Mikolajczak SA, Sattabongkot J, Patrapuvich R, Lindner S, Flannery EL, Koepfli C, Ansell B, Lerch A, Emery-Corbin SJ, Charnaud S, Smith J, Merrienne N, Swearingen KE, Moritz RL, Petter M, Duffy MF, Chuenchob V. Transcriptome and histone epigenome of Plasmodium vivax salivary-gland sporozoites point to tight regulatory control and mechanisms for liver-stage differentiation in relapsing malaria. Int J Parasitol 2019; 49:501-513. [PMID: 31071319 PMCID: PMC9973533 DOI: 10.1016/j.ijpara.2019.02.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 02/01/2019] [Accepted: 02/07/2019] [Indexed: 01/21/2023]
Abstract
Plasmodium vivax is the key obstacle to malaria elimination in Asia and Latin America, largely attributed to its ability to form resilient hypnozoites (sleeper cells) in the host liver that escape treatment and cause relapsing infections. The decision to form hypnozoites is made early in the liver infection and may already be set in sporozoites prior to invasion. To better understand these early stages of infection, we undertook a comprehensive transcriptomic and histone epigenetic characterization of P. vivax sporozoites. Through comparisons with recently published proteomic data for the P. vivax sporozoite, our study found that although highly transcribed, transcripts associated with functions needed for early infection of the vertebrate host are not detectable as proteins and may be regulated through translational repression. We identified differential transcription between the sporozoite and published transcriptomes of asexual blood stages and mixed versus hypnozoite-enriched liver stages. These comparisons point to multiple layers of transcriptional, post-transcriptional and post-translational control that appear active in sporozoites and to a lesser extent hypnozoites, but are largely absent in replicating liver schizonts or mixed blood stages. We also characterised histone epigenetic modifications in the P. vivax sporozoite and explored their role in regulating transcription. Collectively, these data support the hypothesis that the sporozoite is a tightly programmed stage to infect the human host and identify mechanisms for hypnozoite formation that may be further explored in liver stage models.
Collapse
Affiliation(s)
| | - Ivo Muller
- Population Health and Immunity Division, The Walter and
Eliza Hall Institute for Medical Research, 1G Royal Parade, Parkville, Victoria,
3052, Australia,Malaria: Parasites & Hosts Unit, Institut Pasteur, 28
Rue de Dr. Roux, 75015, Paris, France,Department of Medical Biology, The University of Melbourne,
Victoria, 3010, Australia
| | - Aaron R. Jex
- Population Health and Immunity Division, The Walter and
Eliza Hall Institute for Medical Research, 1G Royal Parade, Parkville, Victoria,
3052, Australia,Department of Medical Biology, The University of Melbourne,
Victoria, 3010, Australia,Faculty of Veterinary and Agricultural Sciences, The
University of Melbourne, Corner of Park and Flemington Road, Parkville, Victoria,
3010, Australia
| | - Stefan H. I. Kappe
- Seattle Children’s Research Institute, 307 Westlake
Avenue North, Suite 500, Seattle, WA 98109, USA
| | - Sebastian A. Mikolajczak
- Seattle Children’s Research Institute, 307 Westlake
Avenue North, Suite 500, Seattle, WA 98109, USA
| | - Jetsumon Sattabongkot
- Mahidol Vivax Research Center, Faculty of Tropical
Medicine, Mahidol University, Bangkok 10400, Thailand
| | | | - Scott Lindner
- Department of Biochemistry and Molecular Biology, Center
for Malaria Research, Pennsylvania State University, University Park, PA 16802,
USA
| | - Erika L. Flannery
- Seattle Children’s Research Institute, 307 Westlake
Avenue North, Suite 500, Seattle, WA 98109, USA
| | - Cristian Koepfli
- Population Health and Immunity Division, The Walter and
Eliza Hall Institute for Medical Research, 1G Royal Parade, Parkville, Victoria,
3052, Australia
| | - Brendan Ansell
- Faculty of Veterinary and Agricultural Sciences, The
University of Melbourne, Corner of Park and Flemington Road, Parkville, Victoria,
3010, Australia
| | - Anita Lerch
- Population Health and Immunity Division, The Walter and
Eliza Hall Institute for Medical Research, 1G Royal Parade, Parkville, Victoria,
3052, Australia
| | - Samantha J Emery-Corbin
- Population Health and Immunity Division, The Walter and
Eliza Hall Institute for Medical Research, 1G Royal Parade, Parkville, Victoria,
3052, Australia
| | - Sarah Charnaud
- Population Health and Immunity Division, The Walter and
Eliza Hall Institute for Medical Research, 1G Royal Parade, Parkville, Victoria,
3052, Australia
| | - Jeffrey Smith
- Population Health and Immunity Division, The Walter and
Eliza Hall Institute for Medical Research, 1G Royal Parade, Parkville, Victoria,
3052, Australia
| | - Nicolas Merrienne
- Malaria: Parasites & Hosts Unit, Institut Pasteur, 28
Rue de Dr. Roux, 75015, Paris, France
| | | | | | - Michaela Petter
- Department of Medicine Royal Melbourne Hospital, The Peter
Doherty Institute, The University of Melbourne, 792 Elizabeth Street, Melbourne,
Victoria 3000, Australia,Institute of Microbiology, University Hospital Erlangen,
Erlangen 91054, Germany
| | - Michael F. Duffy
- Department of Medicine Royal Melbourne Hospital, The Peter
Doherty Institute, The University of Melbourne, 792 Elizabeth Street, Melbourne,
Victoria 3000, Australia
| | - Vorada Chuenchob
- Seattle Children’s Research Institute, 307 Westlake
Avenue North, Suite 500, Seattle, WA 98109, USA
| |
Collapse
|