1
|
Miyazaki Y, Miyazaki S. Reporter parasite lines: valuable tools for the study of Plasmodium biology. Trends Parasitol 2024; 40:1000-1015. [PMID: 39389901 DOI: 10.1016/j.pt.2024.09.003] [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: 07/24/2024] [Revised: 08/30/2024] [Accepted: 09/09/2024] [Indexed: 10/12/2024]
Abstract
The human malaria parasite Plasmodium falciparum causes the most severe form of malaria in endemic regions and is transmitted via mosquito bites. To better understand the biology of this deadly pathogen, a variety of P. falciparum reporter lines have been generated using transgenic approaches to express reporter proteins, such as fluorescent proteins and luciferases. This review discusses the advances in recently generated P. falciparum transgenic reporter lines, which will aid in the investigation of parasite physiology and the discovery of novel antimalarial drugs. Future prospects for the generation of new and superior human malaria parasite reporter lines are also discussed, and unresolved questions in malaria biology are highlighted to help boost support for the development and implementation of malaria treatments.
Collapse
Affiliation(s)
- Yukiko Miyazaki
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Japan
| | - Shinya Miyazaki
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Japan.
| |
Collapse
|
2
|
Dirkx L, Van Acker SI, Nicolaes Y, Cunha JLR, Ahmad R, Hendrickx R, Caljon B, Imamura H, Ebo DG, Jeffares DC, Sterckx YGJ, Maes L, Hendrickx S, Caljon G. Long-term hematopoietic stem cells trigger quiescence in Leishmania parasites. PLoS Pathog 2024; 20:e1012181. [PMID: 38656959 PMCID: PMC11073788 DOI: 10.1371/journal.ppat.1012181] [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: 09/05/2023] [Revised: 05/06/2024] [Accepted: 04/09/2024] [Indexed: 04/26/2024] Open
Abstract
Addressing the challenges of quiescence and post-treatment relapse is of utmost importance in the microbiology field. This study shows that Leishmania infantum and L. donovani parasites rapidly enter into quiescence after an estimated 2-3 divisions in both human and mouse bone marrow stem cells. Interestingly, this behavior is not observed in macrophages, which are the primary host cells of the Leishmania parasite. Transcriptional comparison of the quiescent and non-quiescent metabolic states confirmed the overall decrease of gene expression as a hallmark of quiescence. Quiescent amastigotes display a reduced size and signs of a rapid evolutionary adaptation response with genetic alterations. Our study provides further evidence that this quiescent state significantly enhances resistance to treatment. Moreover, transitioning through quiescence is highly compatible with sand fly transmission and increases the potential of parasites to infect cells. Collectively, this work identified stem cells in the bone marrow as a niche where Leishmania quiescence occurs, with important implications for antiparasitic treatment and acquisition of virulence traits.
Collapse
Affiliation(s)
- Laura Dirkx
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Sara I. Van Acker
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Yasmine Nicolaes
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - João Luís Reis Cunha
- York Biomedical Research Institute and Department of Biology, University of York, York, United Kingdom
| | - Rokaya Ahmad
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Rik Hendrickx
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Ben Caljon
- Brussels Interuniversity Genomics High Throughput core (BRIGHTcore) platform, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Hideo Imamura
- Brussels Interuniversity Genomics High Throughput core (BRIGHTcore) platform, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Didier G. Ebo
- Department of Immunology–Allergology–Rheumatology, Faculty of Medicine and Health Science, Infla-Med Centre of Excellence, University of Antwerp, Antwerp University Hospital, Antwerp, Belgium
| | - Daniel C. Jeffares
- York Biomedical Research Institute and Department of Biology, University of York, York, United Kingdom
| | - Yann G.-J. Sterckx
- Laboratory of Medical Biochemistry (LMB), Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Louis Maes
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Sarah Hendrickx
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Guy Caljon
- Laboratory of Microbiology, Parasitology and Hygiene (LMPH), Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
3
|
Mitchell G, Torres L, Fishbaugher ME, Lam M, Chuenchob V, Zalpuri R, Ramasubban S, Baxter CN, Flannery EL, Harupa A, Mikolajczak SA, Jorgens DM. Correlative light-electron microscopy methods to characterize the ultrastructural features of the replicative and dormant liver stages of Plasmodium parasites. Malar J 2024; 23:53. [PMID: 38383417 PMCID: PMC10882739 DOI: 10.1186/s12936-024-04862-w] [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: 12/03/2023] [Accepted: 01/25/2024] [Indexed: 02/23/2024] Open
Abstract
BACKGROUND The infection of the liver by Plasmodium parasites is an obligatory step leading to malaria disease. Following hepatocyte invasion, parasites differentiate into replicative liver stage schizonts and, in the case of Plasmodium species causing relapsing malaria, into hypnozoites that can lie dormant for extended periods of time before activating. The liver stages of Plasmodium remain elusive because of technical challenges, including low infection rate. This has been hindering experimentations with well-established technologies, such as electron microscopy. A deeper understanding of hypnozoite biology could prove essential in the development of radical cure therapeutics against malaria. RESULTS The liver stages of the rodent parasite Plasmodium berghei, causing non-relapsing malaria, and the simian parasite Plasmodium cynomolgi, causing relapsing malaria, were characterized in human Huh7 cells or primary non-human primate hepatocytes using Correlative Light-Electron Microscopy (CLEM). Specifically, CLEM approaches that rely on GFP-expressing parasites (GFP-CLEM) or on an immunofluorescence assay (IFA-CLEM) were used for imaging liver stages. The results from P. berghei showed that host and parasite organelles can be identified and imaged at high resolution using both CLEM approaches. While IFA-CLEM was associated with more pronounced extraction of cellular content, samples' features were generally well preserved. Using IFA-CLEM, a collection of micrographs was acquired for P. cynomolgi liver stage schizonts and hypnozoites, demonstrating the potential of this approach for characterizing the liver stages of Plasmodium species causing relapsing malaria. CONCLUSIONS A CLEM approach that does not rely on parasites expressing genetically encoded tags was developed, therefore suitable for imaging the liver stages of Plasmodium species that lack established protocols to perform genetic engineering. This study also provides a dataset that characterizes the ultrastructural features of liver stage schizonts and hypnozoites from the simian parasite species P. cynomolgi.
Collapse
Affiliation(s)
- Gabriel Mitchell
- Open Innovation at Global Health Disease Area, Biomedical Research, Novartis, Emeryville, CA, USA.
| | - Laura Torres
- Open Innovation at Global Health Disease Area, Biomedical Research, Novartis, Emeryville, CA, USA
| | | | - Melanie Lam
- Open Innovation at Global Health Disease Area, Biomedical Research, Novartis, Emeryville, CA, USA
| | - Vorada Chuenchob
- Global Health Disease Area, Biomedical Research, Novartis, Emeryville, CA, USA
| | - Reena Zalpuri
- Electron Microscope Laboratory, University of California, Berkeley, CA, USA
| | - Shreya Ramasubban
- Electron Microscope Laboratory, University of California, Berkeley, CA, USA
| | - Caitlin N Baxter
- Electron Microscope Laboratory, University of California, Berkeley, CA, USA
| | - Erika L Flannery
- Global Health Disease Area, Biomedical Research, Novartis, Emeryville, CA, USA
| | - Anke Harupa
- Global Health Disease Area, Biomedical Research, Novartis, Emeryville, CA, USA
| | | | - Danielle M Jorgens
- Electron Microscope Laboratory, University of California, Berkeley, CA, USA
| |
Collapse
|
4
|
Voorberg-van der Wel A, Zeeman AM, Kocken CHM. Transfection Models to Investigate Plasmodium vivax-Type Dormant Liver Stage Parasites. Pathogens 2023; 12:1070. [PMID: 37764878 PMCID: PMC10534883 DOI: 10.3390/pathogens12091070] [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: 07/18/2023] [Revised: 08/17/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023] Open
Abstract
Plasmodium vivax causes the second highest number of malaria morbidity and mortality cases in humans. Several biological traits of this parasite species, including the formation of dormant stages (hypnozoites) that persist inside the liver for prolonged periods of time, present an obstacle for intervention measures and create a barrier for the elimination of malaria. Research into the biology of hypnozoites requires efficient systems for parasite transmission, liver stage cultivation and genetic modification. However, P. vivax research is hampered by the lack of an in vitro blood stage culture system, rendering it reliant on in vivo-derived, mainly patient, material for transmission and liver stage culture. This has also resulted in limited capability for genetic modification, creating a bottleneck in investigations into the mechanisms underlying the persistence of the parasite inside the liver. This bottleneck can be overcome through optimal use of the closely related and experimentally more amenable nonhuman primate (NHP) parasite, Plasmodium cynomolgi, as a model system. In this review, we discuss the genetic modification tools and liver stage cultivation platforms available for studying P. vivax persistent stages and highlight how their combined use may advance our understanding of hypnozoite biology.
Collapse
Affiliation(s)
- Annemarie Voorberg-van der Wel
- Department of Parasitology, Biomedical Primate Research Centre, 2288 GJ Rijswijk, The Netherlands; (A.-M.Z.); (C.H.M.K.)
| | | | | |
Collapse
|
5
|
Miyazaki Y, Vos MW, Geurten FJA, Bigeard P, Kroeze H, Yoshioka S, Arisawa M, Inaoka DK, Soulard V, Dechering KJ, Franke-Fayard B, Miyazaki S. A versatile Plasmodium falciparum reporter line expressing NanoLuc enables highly sensitive multi-stage drug assays. Commun Biol 2023; 6:713. [PMID: 37438491 DOI: 10.1038/s42003-023-05078-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 06/27/2023] [Indexed: 07/14/2023] Open
Abstract
Transgenic luciferase-expressing Plasmodium falciparum parasites have been widely used for the evaluation of anti-malarial compounds. Here, to screen for anti-malarial drugs effective against multiple stages of the parasite, we generate a P. falciparum reporter parasite that constitutively expresses NanoLuciferase (NanoLuc) throughout its whole life cycle. The NanoLuc-expressing P. falciparum reporter parasite shows a quantitative NanoLuc signal in the asexual blood, gametocyte, mosquito, and liver stages. We also establish assay systems to evaluate the anti-malarial activity of compounds at the asexual blood, gametocyte, and liver stages, and then determine the 50% inhibitory concentration (IC50) value of several anti-malarial compounds. Through the development of this robust high-throughput screening system, we identify an anti-malarial compound that kills the asexual blood stage parasites. Our study highlights the utility of the NanoLuc reporter line, which may advance anti-malarial drug development through the improved screening of compounds targeting the human malarial parasite at multiple stages.
Collapse
Affiliation(s)
- Yukiko Miyazaki
- Department of Molecular Infection Dynamics, Institute of Tropical Medicine (NEKKEN), Nagasaki University, 852-8523, Nagasaki, Japan.
- Department of Parasitology, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands.
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan.
| | - Martijn W Vos
- TropIQ Health Sciences, Transistorweg 5, 6534 AT, Nijmegen, The Netherlands
| | - Fiona J A Geurten
- Department of Parasitology, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands
| | - Pierre Bigeard
- Sorbonne Université, Inserm, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, F-75013, Paris, France
| | - Hans Kroeze
- Department of Parasitology, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands
| | - Shohei Yoshioka
- Graduate School of Pharmaceutical Sciences, Osaka University, 565-0871, Osaka, Japan
| | - Mitsuhiro Arisawa
- Graduate School of Pharmaceutical Sciences, Osaka University, 565-0871, Osaka, Japan
| | - Daniel Ken Inaoka
- Department of Molecular Infection Dynamics, Institute of Tropical Medicine (NEKKEN), Nagasaki University, 852-8523, Nagasaki, Japan
- School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki, 852-8523, Japan
- Department of Biomedical Chemistry, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Valerie Soulard
- Sorbonne Université, Inserm, CNRS, Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris, F-75013, Paris, France
| | - Koen J Dechering
- TropIQ Health Sciences, Transistorweg 5, 6534 AT, Nijmegen, The Netherlands
| | - Blandine Franke-Fayard
- Department of Parasitology, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands
| | - Shinya Miyazaki
- Department of Parasitology, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands.
- Department of Cellular Architecture Studies, Institute of Tropical Medicine (NEKKEN), Nagasaki University, 852-8523, Nagasaki, Japan.
| |
Collapse
|
6
|
Mitchell G, Roma G, Voorberg-van der Wel A, Beibel M, Zeeman AM, Schuierer S, Torres L, Flannery EL, Kocken CHM, Mikolajczak SA, Diagana TT. Transcriptional profiling of hepatocytes infected with the replicative form of the malaria parasite Plasmodium cynomolgi. Malar J 2022; 21:393. [PMID: 36564750 PMCID: PMC9789591 DOI: 10.1186/s12936-022-04411-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 12/10/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The zoonotic simian parasite Plasmodium cynomolgi develops into replicating schizonts and dormant hypnozoites during the infection of hepatocytes and is used as a model organism to study relapsing malaria. The transcriptional profiling of P. cynomolgi liver stages was previously reported and revealed many important biological features of the parasite but left out the host response to malaria infection. METHODS Previously published RNA sequencing data were used to quantify the expression of host genes in rhesus macaque hepatocytes infected with P. cynomolgi in comparison to either cells from uninfected samples or uninfected bystander cells. RESULTS Although the dataset could not be used to resolve the transcriptional profile of hypnozoite-infected hepatocytes, it provided a snapshot of the host response to liver stage schizonts at 9-10 day post-infection and identified specific host pathways that are modulated during the exo-erythrocytic stage of P. cynomolgi. CONCLUSIONS This study constitutes a valuable resource characterizing the hepatocyte response to P. cynomolgi infection and provides a framework to build on future research that aims at understanding hepatocyte-parasite interactions during relapsing malaria infection.
Collapse
Affiliation(s)
- Gabriel Mitchell
- Open Innovation at Novartis Institute for Tropical Diseases, Novartis Institutes for BioMedical Research, Emeryville, CA, USA.
| | - Guglielmo Roma
- Chemical Biology & Therapeutics, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | | | - Martin Beibel
- Chemical Biology & Therapeutics, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Anne-Marie Zeeman
- Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - Sven Schuierer
- Chemical Biology & Therapeutics, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Laura Torres
- Open Innovation at Novartis Institute for Tropical Diseases, Novartis Institutes for BioMedical Research, Emeryville, CA, USA
| | - Erika L Flannery
- Novartis Institute for Tropical Diseases, Novartis Institutes for BioMedical Research, Emeryville, CA, USA
| | - Clemens H M Kocken
- Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - Sebastian A Mikolajczak
- Novartis Institute for Tropical Diseases, Novartis Institutes for BioMedical Research, Emeryville, CA, USA
| | - Thierry T Diagana
- Novartis Institute for Tropical Diseases, Novartis Institutes for BioMedical Research, Emeryville, CA, USA
| |
Collapse
|
7
|
Sterile protection against relapsing malaria with a single-shot vaccine. NPJ Vaccines 2022; 7:126. [PMID: 36302860 DOI: 10.1038/s41541-022-00555-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 10/11/2022] [Indexed: 11/08/2022] Open
Abstract
Vaccine development for Plasmodium vivax, an important human relapsing malaria, is lagging behind. In the case of the most deadly human malaria P. falciparum, unprecedented high levels of protection have been obtained by immunization with live sporozoites under accompanying chemoprophylaxis, which prevents the onset of blood-stage malaria. Such an approach has not been fully evaluated for relapsing malaria. Here, in the P. cynomolgi-rhesus macaque model for relapsing malaria, we employ the parasites' natural relapsing phenotype to self-boost the immune response against liver-stage parasites, following a single-shot high-dose live sporozoite vaccination. This approach resulted in sterile protection against homologous sporozoite challenge in three out of four animals in the group that was also exposed for several days to blood stages during primary infection and relapses. One out of four animals in the group that received continuous chemoprophylaxis to abort blood-stage exposure was also protected from sporozoite challenge. Although obtained in a small number of animals as part of a Proof-of-Concept study, these results suggest that limited blood-stage parasite exposure may augment protection in this model. We anticipate our data are a starting point for further research into correlates of protection and extrapolation of the single-shot approach to develop efficacious malaria vaccines against relapsing human malaria.
Collapse
|
8
|
Botnar A, Lawrence G, Maher SP, Vantaux A, Witkowski B, Shiau JC, Merino EF, De Vore D, Yang C, Murray C, Cassera MB, Leahy JW, Kyle DE. Alkyne modified purines for assessment of activation of Plasmodium vivax hypnozoites and growth of pre-erythrocytic and erythrocytic stages in Plasmodium spp. Int J Parasitol 2022; 52:733-744. [PMID: 35447149 PMCID: PMC9576819 DOI: 10.1016/j.ijpara.2022.03.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 03/28/2022] [Accepted: 03/28/2022] [Indexed: 12/26/2022]
Abstract
Malaria is a major global health problem which predominantly afflicts developing countries. Although many antimalarial therapies are currently available, the protozoan parasite causing this disease, Plasmodium spp., continues to evade eradication efforts. One biological phenomenon hampering eradication efforts is the parasite's ability to arrest development, transform into a drug-insensitive form, and then resume growth post-therapy. Currently, the mechanisms by which the parasite enters arrested development, or dormancy, and later recrudesces or reactivates to continue development, are unknown and the malaria field lacks techniques to study these elusive mechanisms. Since Plasmodium spp. salvage purines for DNA synthesis, we hypothesised that alkyne-containing purine nucleosides could be used to develop a DNA synthesis marker which could be used to investigate mechanisms behind dormancy. Using copper-catalysed click chemistry methods, we observe incorporation of alkyne modified adenosine, inosine, and hypoxanthine in actively replicating asexual blood stages of Plasmodium falciparum and incorporation of modified adenosine in actively replicating liver stage schizonts of Plasmodium vivax. Notably, these modified purines were not incorporated in dormant liver stage hypnozoites, suggesting this marker could be used as a tool to differentiate replicating and non-replicating liver forms and, more broadly, as a tool for advancing our understanding of Plasmodium dormancy mechanisms.
Collapse
Affiliation(s)
- Alona Botnar
- Department of Infectious Diseases, University of Georgia, Athens, GA, United States; Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, United States
| | - Grant Lawrence
- Department of Chemistry, University of South Florida, Tampa, FL, United States
| | - Steven P Maher
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, United States
| | - Amélie Vantaux
- Malaria Molecular Epidemiology Unit, Pasteur Institute of Cambodia, Phnom Penh, Cambodia
| | - Benoît Witkowski
- Malaria Molecular Epidemiology Unit, Pasteur Institute of Cambodia, Phnom Penh, Cambodia
| | - Justine C Shiau
- Department of Infectious Diseases, University of Georgia, Athens, GA, United States; Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, United States
| | - Emilio F Merino
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, United States; Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, United States
| | - David De Vore
- Department of Chemistry, University of South Florida, Tampa, FL, United States
| | - Christian Yang
- Department of Chemistry, University of South Florida, Tampa, FL, United States
| | - Cameron Murray
- Department of Chemistry, University of South Florida, Tampa, FL, United States
| | - Maria B Cassera
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, United States; Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, United States
| | - James W Leahy
- Department of Chemistry, University of South Florida, Tampa, FL, United States; The Florida Center for Excellence for Drug Discovery and Innovation, University of South Florida, Tampa, GA, United States; Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, GA, United States
| | - Dennis E Kyle
- Department of Infectious Diseases, University of Georgia, Athens, GA, United States; Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, United States; Department of Cellular Biology, University of Georgia, Athens, GA, United States.
| |
Collapse
|
9
|
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
|
10
|
Simwela NV, Waters AP. Current status of experimental models for the study of malaria. Parasitology 2022; 149:1-22. [PMID: 35357277 PMCID: PMC9378029 DOI: 10.1017/s0031182021002134] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 01/09/2023]
Abstract
Infection by malaria parasites (Plasmodium spp.) remains one of the leading causes of morbidity and mortality, especially in tropical regions of the world. Despite the availability of malaria control tools such as integrated vector management and effective therapeutics, these measures have been continuously undermined by the emergence of vector resistance to insecticides or parasite resistance to frontline antimalarial drugs. Whilst the recent pilot implementation of the RTS,S malaria vaccine is indeed a remarkable feat, highly effective vaccines against malaria remain elusive. The barriers to effective vaccines result from the complexity of both the malaria parasite lifecycle and the parasite as an organism itself with consequent major gaps in our understanding of their biology. Historically and due to the practical and ethical difficulties of working with human malaria infections, research into malaria parasite biology has been extensively facilitated by animal models. Animals have been used to study disease pathogenesis, host immune responses and their (dys)regulation and further disease processes such as transmission. Moreover, animal models remain at the forefront of pre-clinical evaluations of antimalarial drugs (drug efficacy, mode of action, mode of resistance) and vaccines. In this review, we discuss commonly used animal models of malaria, the parasite species used and their advantages and limitations which hinder their extrapolation to actual human disease. We also place into this context the most recent developments such as organoid technologies and humanized mice.
Collapse
Affiliation(s)
- Nelson V. Simwela
- Institute of Infection, Immunity & Inflammation, Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
| | - Andrew P. Waters
- Institute of Infection, Immunity & Inflammation, Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
| |
Collapse
|
11
|
Vieira TB, Astro TP, de Moraes Barros RR. Genetic Manipulation of Non-Falciparum Human Malaria Parasites. Front Cell Infect Microbiol 2021; 11:680460. [PMID: 34527600 PMCID: PMC8435838 DOI: 10.3389/fcimb.2021.680460] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 08/11/2021] [Indexed: 11/13/2022] Open
Abstract
The development of genetic manipulation of Plasmodium falciparum in the 1980s was key to study malaria biology. Genetically modified parasites have been used to study several aspects of the disease, such as red blood cell invasion, drug resistance mechanisms, gametocyte development and mosquito transmission. However, biological and genetic differences between P. falciparum and the other human malaria parasites make P. falciparum a poor model to study different species. The lack of robust systems of long-term in vitro culture of P. vivax and the other human malaria parasites lagged the genetic manipulation of these species. Here we review the efforts to generate genetically modified non-falciparum human malaria parasites, in vivo and in vitro. Using in vivo models – infection of non-human primates such as rhesus macaques and saimiri monkeys – researchers were able to generate transgenic lines of P. knowlesi, P. cynomolgi, and P. vivax. The development of long-term in vitro culture of P. knowlesi in the 2000’s, using rhesus and human red blood cells, created a platform to genetically manipulate non-falciparum malaria parasites. Recently, the use of CRISPR/Cas9 technology to genome edit P. knowlesi provides another tool to non-falciparum malaria research, extending the possibilities and allowing researchers to study different aspects of the biology of these parasites and understand the differences between these species and P. falciparum.
Collapse
Affiliation(s)
- Taís Baruel Vieira
- Department of Microbiology, Immunology and Parasitology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Thafne Plastina Astro
- Department of Microbiology, Immunology and Parasitology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Roberto Rudge de Moraes Barros
- Department of Microbiology, Immunology and Parasitology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
12
|
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
|
13
|
Sylvester K, Maher SP, Posfai D, Tran MK, Crawford MC, Vantaux A, Witkowski B, Kyle DE, Derbyshire ER. Characterization of the Tubovesicular Network in Plasmodium vivax Liver Stage Hypnozoites and Schizonts. Front Cell Infect Microbiol 2021; 11:687019. [PMID: 34195101 PMCID: PMC8236947 DOI: 10.3389/fcimb.2021.687019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 05/24/2021] [Indexed: 12/04/2022] Open
Abstract
Plasmodium is a genus of apicomplexan parasites which replicate in the liver before causing malaria. Plasmodium vivax can also persist in the liver as dormant hypnozoites and cause clinical relapse upon activation, but the molecular mechanisms leading to activation have yet to be discovered. In this study, we use high-resolution microscopy to characterize temporal changes of the P. vivax liver stage tubovesicular network (TVN), a parasitophorous vacuole membrane (PVM)-derived network within the host cytosol. We observe extended membrane clusters, tubules, and TVN-derived vesicles present throughout P. vivax liver stage development. Additionally, we demonstrate an unexpected presence of the TVN in hypnozoites and observe some association of this network to host nuclei. We also reveal that the host water and solute channel aquaporin-3 (AQP3) associates with TVN-derived vesicles and extended membrane clusters. AQP3 has been previously shown to localize to the PVM of P. vivax hypnozoites and liver schizonts but has not yet been shown in association to the TVN. Our results highlight host-parasite interactions occur in both dormant and replicating liver stage P. vivax forms and implicate AQP3 function during this time. Together, these findings enhance our understanding of P. vivax liver stage biology through characterization of the TVN with an emphasis on the presence of this network in dormant hypnozoites.
Collapse
Affiliation(s)
- Kayla Sylvester
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, United States
| | - Steven P Maher
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, United States
| | - Dora Posfai
- Chemistry Department, Duke University, Durham, NC, United States
| | - Michael K Tran
- Chemistry Department, Duke University, Durham, NC, United States
| | | | - Amélie Vantaux
- Malaria Molecular Epidemiology Unit, Pasteur Institute in Cambodia, Phnom Penh, Cambodia
| | - Benoît Witkowski
- Malaria Molecular Epidemiology Unit, Pasteur Institute in Cambodia, Phnom Penh, Cambodia
| | - Dennis E Kyle
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, United States
| | - Emily R Derbyshire
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, United States.,Chemistry Department, Duke University, Durham, NC, United States
| |
Collapse
|
14
|
Moraes Barros RR, Thawnashom K, Gibson TJ, Armistead JS, Caleon RL, Kaneko M, Kite WA, Mershon JP, Brockhurst JK, Engels T, Lambert L, Orr-Gonzalez S, Adams JH, Sá JM, Kaneko O, Wellems TE. Activity of Plasmodium vivax promoter elements in Plasmodium knowlesi, and a centromere-containing plasmid that expresses NanoLuc throughout the parasite life cycle. Malar J 2021; 20:247. [PMID: 34090438 PMCID: PMC8180018 DOI: 10.1186/s12936-021-03773-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 05/16/2021] [Indexed: 12/23/2022] Open
Abstract
Background Plasmodium knowlesi is now the major cause of human malaria in Malaysia, complicating malaria control efforts that must attend to the elimination of multiple Plasmodium species. Recent advances in the cultivation of P. knowlesi erythrocytic-stage parasites in vitro, transformation with exogenous DNA, and infection of mosquitoes with gametocytes from culture have opened up studies of this pathogen without the need for resource-intensive and costly non-human primate (NHP) models. For further understanding and development of methods for parasite transformation in malaria research, this study examined the activity of various trans-species transcriptional control sequences and the influence of Plasmodium vivax centromeric (pvcen) repeats in plasmid-transfected P. knowlesi parasites. Methods In vitro cultivated P. knowlesi parasites were transfected with plasmid constructs that incorporated Plasmodium vivax or Plasmodium falciparum 5′ UTRs driving the expression of bioluminescence markers (firefly luciferase or Nanoluc). Promoter activities were assessed by bioluminescence, and parasites transformed with human resistant allele dihydrofolate reductase-expressing plasmids were selected using antifolates. The stability of transformants carrying pvcen-stabilized episomes was assessed by bioluminescence over a complete parasite life cycle through a rhesus macaque monkey, mosquitoes, and a second rhesus monkey. Results Luciferase expression assessments show that certain P. vivax promoter regions, not functional in the more evolutionarily-distant P. falciparum, can drive transgene expression in P. knowlesi. Further, pvcen repeats may improve the stability of episomal plasmids in P. knowlesi and support detection of NanoLuc-expressing elements over the full parasite life cycle from rhesus macaque monkeys to Anopheles dirus mosquitoes and back again to monkeys. In assays of drug responses to chloroquine, G418 and WR9910, anti-malarial half-inhibitory concentration (IC50) values of blood stages measured by NanoLuc activity proved comparable to IC50 values measured by the standard SYBR Green method. Conclusion All three P. vivax promoters tested in this study functioned in P. knowlesi, whereas two of the three were inactive in P. falciparum. NanoLuc-expressing, centromere-stabilized plasmids may support high-throughput screenings of P. knowlesi for new anti-malarial agents, including compounds that can block the development of mosquito- and/or liver-stage parasites. Supplementary Information The online version contains supplementary material available at 10.1186/s12936-021-03773-4.
Collapse
Affiliation(s)
- Roberto R Moraes Barros
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA. .,Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil.
| | - Kittisak Thawnashom
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan.,Department of Medical Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, Thailand
| | - Tyler J Gibson
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jennifer S Armistead
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.,Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, FL, USA
| | - Ramoncito L Caleon
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Miho Kaneko
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan.,Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Whitney A Kite
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - J Patrick Mershon
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jacqueline K Brockhurst
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Theresa Engels
- Division of Veterinary Research, National Institutes of Health, Bethesda, MD, USA
| | - Lynn Lambert
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sachy Orr-Gonzalez
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - John H Adams
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, FL, USA
| | - Juliana M Sá
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Osamu Kaneko
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
| | - Thomas E Wellems
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
15
|
der Wel AVV, Hofman SO, Kocken CHM. Isolation of GFP-expressing Malarial Hypnozoites by Flow Cytometry Cell Sorting. Bio Protoc 2021; 11:e4006. [PMID: 34124306 DOI: 10.21769/bioprotoc.4006] [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: 11/23/2020] [Revised: 02/14/2021] [Accepted: 03/19/2021] [Indexed: 11/02/2022] Open
Abstract
Hypnozoites are dormant liver-stage parasites unique to relapsing malarial species, including the important human pathogen Plasmodium vivax, and pose a barrier to the elimination of malaria. Little is known regarding the biology of these stages, largely due to their inaccessible location. Hypnozoites can be cultured in vitro but these cultures always consist of a mixture of hepatocytes, developing forms, and hypnozoites. Here, using a GFP-expressing line of the hypnozoite model parasite Plasmodium cynomolgi, we describe a protocol for the FACS-based isolation of malarial hypnozoites. The purified hypnozoites can be used for a range of '-omics' studies to dissect the biology of this cryptic stage of the malarial life cycle.
Collapse
Affiliation(s)
| | - Sam O Hofman
- Department of Parasitology, Biomedical Primate Research Centre, 2288 GJ Rijswijk, The Netherlands
| | - Clemens H M Kocken
- Department of Parasitology, Biomedical Primate Research Centre, 2288 GJ Rijswijk, The Netherlands
| |
Collapse
|
16
|
Buyon LE, Elsworth B, Duraisingh MT. The molecular basis of antimalarial drug resistance in Plasmodium vivax. INTERNATIONAL JOURNAL FOR PARASITOLOGY-DRUGS AND DRUG RESISTANCE 2021; 16:23-37. [PMID: 33957488 PMCID: PMC8113647 DOI: 10.1016/j.ijpddr.2021.04.002] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 03/31/2021] [Accepted: 04/08/2021] [Indexed: 01/07/2023]
Abstract
Plasmodium vivax is the most geographically widespread cause of human malaria and is responsible for the majority of cases outside of the African continent. While great progress has been made towards eliminating human malaria, drug resistant parasite strains pose a threat towards continued progress. Resistance has arisen to multiple antimalarials in P. vivax, including to chloroquine, which is currently the first line therapy for P. vivax in most regions. Despite its importance, an understanding of the molecular mechanisms of drug resistance in this species remains elusive, in large part due to the complex biology of P. vivax and the lack of in vitro culture. In this review, we will cover the extent and challenges of measuring clinical and in vitro drug resistance in P. vivax. We will consider the roles of candidate drug resistance genes. We will highlight the development of molecular approaches for studying P. vivax biology that provide the opportunity to validate the role of putative drug resistance mutations as well as identify novel mechanisms of drug resistance in this understudied parasite. Validated molecular determinants and markers of drug resistance are essential for the rapid and cost-effective monitoring of drug resistance in P. vivax, and will be useful for optimizing drug regimens and for informing drug policy in control and elimination settings. Drug resistance is emerging in Plasmodium vivax, an important cause of malaria. The complex biology of P. vivax and the limited range of research tools make it difficult to identify drug resistance. The molecular mechanisms of drug resistance in P. vivax remain elusive. This review highlights the extent of drug resistance, the putative mechanisms of resistance and new technologies for the study of P. vivax drug resistance.
Collapse
Affiliation(s)
- Lucas E Buyon
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, 02115, MA, USA
| | - Brendan Elsworth
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, 02115, MA, USA
| | - Manoj T Duraisingh
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, 02115, MA, USA.
| |
Collapse
|
17
|
The New Zoonotic Malaria: Plasmodium cynomolgi. Trop Med Infect Dis 2021; 6:tropicalmed6020046. [PMID: 33916448 PMCID: PMC8167800 DOI: 10.3390/tropicalmed6020046] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 04/02/2021] [Accepted: 04/03/2021] [Indexed: 12/04/2022] Open
Abstract
Plasmodium cynomolgi is a simian malaria parasite that has been a central model parasite since it was first described in 1907. Recently it has made the zoonotic jump and started naturally infecting humans. In this paper, the interactions between Plasmodium cynomolgi and humans, the environment and the non-human animal intermediates or definitive host will be discussed, with a particular focus on the clinical implications of infection and approaches to management of this novel zoonotic parasite.
Collapse
|
18
|
Voorberg-van der Wel A, Kocken CHM, Zeeman AM. Modeling Relapsing Malaria: Emerging Technologies to Study Parasite-Host Interactions in the Liver. Front Cell Infect Microbiol 2021; 10:606033. [PMID: 33585277 PMCID: PMC7878928 DOI: 10.3389/fcimb.2020.606033] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 12/04/2020] [Indexed: 01/03/2023] Open
Abstract
Recent studies of liver stage malaria parasite-host interactions have provided exciting new insights on the cross-talk between parasite and its mammalian (predominantly rodent) host. We review the latest state of the art and and zoom in on new technologies that will provide the tools necessary to investigate host-parasite interactions of relapsing parasites. Interactions between hypnozoites and hepatocytes are particularly interesting because the parasite can remain in a quiescent state for prolonged periods of time and triggers for reactivation have not been irrefutably identified. If we learn more about the cross-talk between hypnozoite and host we may be able to identify factors that encourage waking up these dormant parasite reservoirs and help to achieve the total eradication of malaria.
Collapse
Affiliation(s)
| | - Clemens H M Kocken
- Department of Parasitology, Biomedical Primate Research Center, Rijswijk, Netherlands
| | - Anne-Marie Zeeman
- Department of Parasitology, Biomedical Primate Research Center, Rijswijk, Netherlands
| |
Collapse
|
19
|
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
|
20
|
Merrick CJ. Hypnozoites in Plasmodium: Do Parasites Parallel Plants? Trends Parasitol 2020; 37:273-282. [PMID: 33257270 DOI: 10.1016/j.pt.2020.11.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/03/2020] [Accepted: 11/03/2020] [Indexed: 12/12/2022]
Abstract
The phenomenon of relapsing malaria has been recognised for centuries. It is caused in humans by the parasite species Plasmodium vivax and Plasmodium ovale, which can arrest growth at an early, asymptomatic stage as hypnozoites inside liver cells. These dormant parasites can remain quiescent for months or years, then reactivate causing symptomatic malaria. The dynamics of hypnozoite dormancy and reactivation are well documented but the molecular basis remains a complete mystery. Here, I observe that the process has striking parallels with plant vernalisation, whereby plants remain dormant through the winter before flowering in spring. Vernalisation is thoroughly studied in several plant species and its mechanisms are known in exquisite detail. Vernalisation may thus provide a useful framework for interrogating hypnozoite biology.
Collapse
Affiliation(s)
- Catherine J Merrick
- Department of Pathology, Cambridge University, Tennis Court Road, Cambridge, CB2 1QP, UK.
| |
Collapse
|
21
|
Voorberg-van der Wel A, Zeeman AM, Nieuwenhuis IG, van der Werff NM, Klooster EJ, Klop O, Vermaat LC, Kocken CHM. Dual-Luciferase-Based Fast and Sensitive Detection of Malaria Hypnozoites for the Discovery of Antirelapse Compounds. Anal Chem 2020; 92:6667-6675. [PMID: 32267675 PMCID: PMC7203758 DOI: 10.1021/acs.analchem.0c00547] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 04/08/2020] [Indexed: 01/24/2023]
Abstract
Efforts to eradicate Plasmodium vivax malaria are hampered by the presence of hypnozoites, persisting stages in the liver that can reactivate after prolonged periods of time enabling further transmission and causing renewed disease. Large-scale drug screening is needed to identify compounds with antihypnozoite activity, but current platforms rely on time-consuming high-content fluorescence imaging as read-out, limiting assay throughput. We here report an ultrafast and sensitive dual-luciferase-based method to differentiate hypnozoites from liver stage schizonts using a transgenic P. cynomolgi parasite line that contains Nanoluc driven by the constitutive hsp70 promoter, as well as firefly luciferase driven by the schizont-specific lisp2 promoter. The transgenic parasite line showed similar fitness and drug sensitivity profiles of selected compounds to wild type. We demonstrate robust bioluminescence-based detection of hypnozoites in 96-well and 384-well plate formats, setting the stage for implementation in large scale drug screens.
Collapse
Affiliation(s)
| | - Anne-Marie Zeeman
- Department of Parasitology, Biomedical Primate Research Centre, 2288 GJ Rijswijk, The Netherlands
| | - Ivonne G. Nieuwenhuis
- Department of Parasitology, Biomedical Primate Research Centre, 2288 GJ Rijswijk, The Netherlands
| | - Nicole M. van der Werff
- Department of Parasitology, Biomedical Primate Research Centre, 2288 GJ Rijswijk, The Netherlands
| | - Els J. Klooster
- Department of Parasitology, Biomedical Primate Research Centre, 2288 GJ Rijswijk, The Netherlands
| | - Onny Klop
- Department of Parasitology, Biomedical Primate Research Centre, 2288 GJ Rijswijk, The Netherlands
| | - Lars C. Vermaat
- Department of Parasitology, Biomedical Primate Research Centre, 2288 GJ Rijswijk, The Netherlands
| | - Clemens H. M. Kocken
- Department of Parasitology, Biomedical Primate Research Centre, 2288 GJ Rijswijk, The Netherlands
| |
Collapse
|
22
|
A dual fluorescent Plasmodium cynomolgi reporter line reveals in vitro malaria hypnozoite reactivation. Commun Biol 2020; 3:7. [PMID: 31909199 PMCID: PMC6941962 DOI: 10.1038/s42003-019-0737-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 12/12/2019] [Indexed: 12/21/2022] Open
Abstract
Plasmodium vivax malaria is characterized by repeated episodes of blood stage infection (relapses) resulting from activation of dormant stages in the liver, so-called hypnozoites. Transition of hypnozoites into developing schizonts has never been observed. A barrier for studying this has been the lack of a system in which to monitor growth of liver stages. Here, exploiting the unique strengths of the simian hypnozoite model P. cynomolgi, we have developed green-fluorescent (GFP) hypnozoites that turn on red-fluorescent (mCherry) upon activation. The transgenic parasites show full liver stage development, including merozoite release and red blood cell infection. We demonstrate that individual hypnozoites actually can activate and resume development after prolonged culture, providing the last missing evidence of the hypnozoite theory of relapse. The few events identified indicate that hypnozoite activation in vitro is infrequent. This system will further our understanding of the mechanisms of hypnozoite activation and may facilitate drug discovery approaches.
Collapse
|
23
|
Thawnashom K, Kaneko M, Xangsayarath P, Chaiyawong N, Yahata K, Asada M, Adams JH, Kaneko O. Validation of Plasmodium vivax centromere and promoter activities using Plasmodium yoelii. PLoS One 2019; 14:e0226884. [PMID: 31860644 PMCID: PMC6924662 DOI: 10.1371/journal.pone.0226884] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 12/06/2019] [Indexed: 11/18/2022] Open
Abstract
Plasmodium vivax is the leading cause of malaria outside Africa and represents a significant health and economic burden on affected countries. A major obstacle for P. vivax eradication is the dormant hypnozoite liver stage that causes relapse infections and the limited antimalarial drugs that clear this stage. Advances in studying the hypnozoite and other unique biological aspects of this parasite are hampered by the lack of a continuous in vitro laboratory culture system and poor availability of molecular tools for genetic manipulation. In this study, we aim to develop molecular tools that can be used for genetic manipulation of P. vivax. A putative P. vivax centromere sequence (PvCEN) was cloned and episomal centromere based plasmids expressing a GFP marker were constructed. Centromere activity was evaluated using a rodent malaria parasite Plasmodium yoelii. A plasmid carrying PvCEN was stably maintained in asexual-stage parasites in the absence of drug pressure, and approximately 45% of the parasites retained the plasmid four weeks later. The same retention rate was observed in parasites possessing a native P. yoelii centromere (PyCEN)-based control plasmid. The segregation efficiency of the plasmid per nuclear division was > 99% in PvCEN parasites, compared to ~90% in a control parasite harboring a plasmid without a centromere. In addition, we observed a clear GFP signal in both oocysts and salivary gland sporozoites isolated from mosquitoes. In blood-stage parasites after liver stage development, GFP positivity in PvCEN parasites was comparable to control PyCEN parasites. Thus, PvCEN plasmids were maintained throughout the parasite life cycle. We also validated several P. vivax promoter activities and showed that hsp70 promoter (~1 kb) was active throughout the parasite life cycle. This is the first data for the functional characterization of a P. vivax centromere that can be used in future P. vivax biological research.
Collapse
Affiliation(s)
- Kittisak Thawnashom
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Sakamoto, Nagasaki, Japan
- Department of Medical Technology, Faculty of Allied Health Sciences, Naresuan University, Mueang, Phitsanulok, Thailand
| | - Miho Kaneko
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Sakamoto, Nagasaki, Japan
| | - Phonepadith Xangsayarath
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Sakamoto, Nagasaki, Japan
| | - Nattawat Chaiyawong
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Sakamoto, Nagasaki, Japan
- Leading Program, Graduate School of Biomedical Sciences, Nagasaki University, Sakamoto, Nagasaki, Japan
| | - Kazuhide Yahata
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Sakamoto, Nagasaki, Japan
| | - Masahito Asada
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Sakamoto, Nagasaki, Japan
- Leading Program, Graduate School of Biomedical Sciences, Nagasaki University, Sakamoto, Nagasaki, Japan
| | - 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
| | - Osamu Kaneko
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Sakamoto, Nagasaki, Japan
- Leading Program, Graduate School of Biomedical Sciences, Nagasaki University, Sakamoto, Nagasaki, Japan
- * E-mail:
| |
Collapse
|
24
|
Barrett MP, Kyle DE, Sibley LD, Radke JB, Tarleton RL. Protozoan persister-like cells and drug treatment failure. Nat Rev Microbiol 2019; 17:607-620. [PMID: 31444481 PMCID: PMC7024564 DOI: 10.1038/s41579-019-0238-x] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/03/2019] [Indexed: 01/01/2023]
Abstract
Antimicrobial treatment failure threatens our ability to control infections. In addition to antimicrobial resistance, treatment failures are increasingly understood to derive from cells that survive drug treatment without selection of genetically heritable mutations. Parasitic protozoa, such as Plasmodium species that cause malaria, Toxoplasma gondii and kinetoplastid protozoa, including Trypanosoma cruzi and Leishmania spp., cause millions of deaths globally. These organisms can evolve drug resistance and they also exhibit phenotypic diversity, including the formation of quiescent or dormant forms that contribute to the establishment of long-term infections that are refractory to drug treatment, which we refer to as 'persister-like cells'. In this Review, we discuss protozoan persister-like cells that have been linked to persistent infections and discuss their impact on therapeutic outcomes following drug treatment.
Collapse
Affiliation(s)
- Michael P Barrett
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK.
| | - Dennis E Kyle
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, USA
| | - L David Sibley
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO, USA
| | - Joshua B Radke
- Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO, USA
| | - Rick L Tarleton
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, USA
| |
Collapse
|
25
|
Norcross NR, Wilson C, Baragaña B, Hallyburton I, Osuna‐Cabello M, Norval S, Riley J, Fletcher D, Sinden R, Delves M, Ruecker A, Duffy S, Meister S, Antonova‐Koch Y, Crespo B, de Cózar C, Sanz LM, Gamo FJ, Avery VM, Frearson JA, Gray DW, Fairlamb AH, Winzeler EA, Waterson D, Campbell SF, Willis PA, Read KD, Gilbert IH. Substituted Aminoacetamides as Novel Leads for Malaria Treatment. ChemMedChem 2019; 14:1329-1335. [PMID: 31188540 PMCID: PMC6899483 DOI: 10.1002/cmdc.201900329] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Indexed: 01/29/2023]
Abstract
Herein we describe the optimization of a phenotypic hit against Plasmodium falciparum based on an aminoacetamide scaffold. This led to N-(3-chloro-4-fluorophenyl)-2-methyl-2-{[4-methyl-3-(morpholinosulfonyl)phenyl]amino}propanamide (compound 28) with low-nanomolar activity against the intraerythrocytic stages of the malaria parasite, and which was found to be inactive in a mammalian cell counter-screen up to 25 μm. Inhibition of gametes in the dual gamete activation assay suggests that this family of compounds may also have transmission blocking capabilities. Whilst we were unable to optimize the aqueous solubility and microsomal stability to a point at which the aminoacetamides would be suitable for in vivo pharmacokinetic and efficacy studies, compound 28 displayed excellent antimalarial potency and selectivity; it could therefore serve as a suitable chemical tool for drug target identification.
Collapse
Affiliation(s)
- Neil R. Norcross
- Drug Discovery UnitDivision of Biological Chemistry and Drug DiscoverySchool of Life SciencesUniversity of DundeeDundeeDD1 5EHUK
| | - Caroline Wilson
- Drug Discovery UnitDivision of Biological Chemistry and Drug DiscoverySchool of Life SciencesUniversity of DundeeDundeeDD1 5EHUK
| | - Beatriz Baragaña
- Drug Discovery UnitDivision of Biological Chemistry and Drug DiscoverySchool of Life SciencesUniversity of DundeeDundeeDD1 5EHUK
| | - Irene Hallyburton
- Drug Discovery UnitDivision of Biological Chemistry and Drug DiscoverySchool of Life SciencesUniversity of DundeeDundeeDD1 5EHUK
| | - Maria Osuna‐Cabello
- Drug Discovery UnitDivision of Biological Chemistry and Drug DiscoverySchool of Life SciencesUniversity of DundeeDundeeDD1 5EHUK
| | - Suzanne Norval
- Drug Discovery UnitDivision of Biological Chemistry and Drug DiscoverySchool of Life SciencesUniversity of DundeeDundeeDD1 5EHUK
| | - Jennifer Riley
- Drug Discovery UnitDivision of Biological Chemistry and Drug DiscoverySchool of Life SciencesUniversity of DundeeDundeeDD1 5EHUK
| | - Daniel Fletcher
- Drug Discovery UnitDivision of Biological Chemistry and Drug DiscoverySchool of Life SciencesUniversity of DundeeDundeeDD1 5EHUK
| | | | | | | | - Sandra Duffy
- Discovery BiologyGriffith Institute for Drug DiscoveryGriffith UniversityNathanQueensland4111Australia
| | - Stephan Meister
- Department of PediatricsUniversity of California San Diego School of Medicine9500 Gilman Drive 0741La JollaCA92093USA
| | - Yevgeniya Antonova‐Koch
- Department of PediatricsUniversity of California San Diego School of Medicine9500 Gilman Drive 0741La JollaCA92093USA
| | - Benigno Crespo
- GlaxoSmithKline, Diseases of the Developing World – Tres Cantos Medicines Development Campusc/ Severo Ochoa 2, Tres Cantos28760MadridSpain
| | - Cristina de Cózar
- GlaxoSmithKline, Diseases of the Developing World – Tres Cantos Medicines Development Campusc/ Severo Ochoa 2, Tres Cantos28760MadridSpain
| | - Laura M. Sanz
- GlaxoSmithKline, Diseases of the Developing World – Tres Cantos Medicines Development Campusc/ Severo Ochoa 2, Tres Cantos28760MadridSpain
| | - Francisco Javier Gamo
- GlaxoSmithKline, Diseases of the Developing World – Tres Cantos Medicines Development Campusc/ Severo Ochoa 2, Tres Cantos28760MadridSpain
| | - Vicky M. Avery
- Discovery BiologyGriffith Institute for Drug DiscoveryGriffith UniversityNathanQueensland4111Australia
| | - Julie A. Frearson
- Drug Discovery UnitDivision of Biological Chemistry and Drug DiscoverySchool of Life SciencesUniversity of DundeeDundeeDD1 5EHUK
| | - David W. Gray
- Drug Discovery UnitDivision of Biological Chemistry and Drug DiscoverySchool of Life SciencesUniversity of DundeeDundeeDD1 5EHUK
| | - Alan H. Fairlamb
- Drug Discovery UnitDivision of Biological Chemistry and Drug DiscoverySchool of Life SciencesUniversity of DundeeDundeeDD1 5EHUK
| | - Elizabeth A. Winzeler
- Department of PediatricsUniversity of California San Diego School of Medicine9500 Gilman Drive 0741La JollaCA92093USA
| | - David Waterson
- Medicines for Malaria VentureInternational Centre, Cointrin, Entrance G, 3rd FloorRoute de Pré-Bois 20, PO Box 1826Geneva1215Switzerland
| | - Simon F. Campbell
- Medicines for Malaria VentureInternational Centre, Cointrin, Entrance G, 3rd FloorRoute de Pré-Bois 20, PO Box 1826Geneva1215Switzerland
| | - Paul A. Willis
- Medicines for Malaria VentureInternational Centre, Cointrin, Entrance G, 3rd FloorRoute de Pré-Bois 20, PO Box 1826Geneva1215Switzerland
| | - Kevin D. Read
- Drug Discovery UnitDivision of Biological Chemistry and Drug DiscoverySchool of Life SciencesUniversity of DundeeDundeeDD1 5EHUK
| | - Ian H. Gilbert
- Drug Discovery UnitDivision of Biological Chemistry and Drug DiscoverySchool of Life SciencesUniversity of DundeeDundeeDD1 5EHUK
| |
Collapse
|
26
|
Chua ACY, Ananthanarayanan A, Ong JJY, Wong JY, Yip A, Singh NH, Qu Y, Dembele L, McMillian M, Ubalee R, Davidson S, Tungtaeng A, Imerbsin R, Gupta K, Andolina C, Lee F, S-W Tan K, Nosten F, Russell B, Lange A, Diagana TT, Rénia L, Yeung BKS, Yu H, Bifani P. Hepatic spheroids used as an in vitro model to study malaria relapse. Biomaterials 2019; 216:119221. [PMID: 31195301 DOI: 10.1016/j.biomaterials.2019.05.032] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 05/19/2019] [Indexed: 12/27/2022]
Abstract
Hypnozoites are the liver stage non-dividing form of the malaria parasite that are responsible for relapse and acts as a natural reservoir for human malaria Plasmodium vivax and P. ovale as well as a phylogenetically related simian malaria P. cynomolgi. Our understanding of hypnozoite biology remains limited due to the technical challenge of requiring the use of primary hepatocytes and the lack of robust and predictive in vitro models. In this study, we developed a malaria liver stage model using 3D spheroid-cultured primary hepatocytes. The infection of primary hepatocytes in suspension led to increased infectivity of both P. cynomolgi and P. vivax infections. We demonstrated that this hepatic spheroid model was capable of maintaining long term viability, hepatocyte specific functions and cell polarity which enhanced permissiveness and thus, permitting for the complete development of both P. cynomolgi and P. vivax liver stage parasites in the infected spheroids. The model described here was able to capture the full liver stage cycle starting with sporozoites and ending in the release of hepatic merozoites capable of invading simian erythrocytes in vitro. Finally, we showed that this system can be used for compound screening to discriminate between causal prophylactic and cidal antimalarials activity in vitro for relapsing malaria.
Collapse
Affiliation(s)
- Adeline C Y Chua
- Novartis Institute for Tropical Diseases, 138670, Singapore; Department of Microbiology and Immunology, University of Otago, Dunedin 9054, New Zealand; Singapore Immunology Network (SIgN), A*STAR, 138648, Singapore
| | | | - Jessica Jie Ying Ong
- Novartis Institute for Tropical Diseases, 138670, Singapore; Department of Microbiology and Immunology, University of Otago, Dunedin 9054, New Zealand
| | | | - Andy Yip
- Novartis Institute for Tropical Diseases, 138670, Singapore
| | | | | | - Laurent Dembele
- Novartis Institute for Tropical Diseases, 138670, Singapore; Université des Sciences, des Techniques et des Technologies de Bamako (USTTB); MRTC - DEAP - Faculty of Pharmacy, Bamako, Mali
| | - Michael McMillian
- Invitrocue Pte Ltd. 138667, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University Health System, 117597, Singapore
| | - Ratawan Ubalee
- Department of Entomology, Armed Forces Research Institute of Medical Science (AFRIMS), Bangkok 10400,Thailand
| | - Silas Davidson
- Department of Entomology, Armed Forces Research Institute of Medical Science (AFRIMS), Bangkok 10400,Thailand
| | - Anchalee Tungtaeng
- Department of Veterinary Medicine, Armed Forces Research Institute of Medical Science (AFRIMS), Bangkok 10400,Thailand
| | - Rawiwan Imerbsin
- Department of Veterinary Medicine, Armed Forces Research Institute of Medical Science (AFRIMS), Bangkok 10400,Thailand
| | - Kapish Gupta
- Mechanobiology Institute, National University of Singapore, 117411, Singapore
| | - Chiara Andolina
- Shoklo Malaria Research Unit (SMRU), Mahidol-Oxford Tropical Medicine 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
| | - Fan Lee
- Institute of Bioengineering and Nanotechnology, A*STAR, 138669, Singapore
| | - Kevin S-W Tan
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 119077, Singapore
| | - François Nosten
- Shoklo Malaria Research Unit (SMRU), Mahidol-Oxford Tropical Medicine 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
| | - Bruce Russell
- Department of Microbiology and Immunology, University of Otago, Dunedin 9054, New Zealand
| | - Amber Lange
- Laboratory Animal Services, Scientific Operations, Novartis Institutes for Biomedical Research, East Hanover, NJ, 07936-1080, USA
| | | | - Laurent Rénia
- Singapore Immunology Network (SIgN), A*STAR, 138648, Singapore; Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 119077, Singapore
| | | | - Hanry Yu
- Invitrocue Pte Ltd. 138667, Singapore; Mechanobiology Institute, National University of Singapore, 117411, Singapore; Institute of Bioengineering and Nanotechnology, A*STAR, 138669, Singapore
| | - Pablo Bifani
- Novartis Institute for Tropical Diseases, 138670, Singapore; Singapore Immunology Network (SIgN), A*STAR, 138648, Singapore; Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 119077, Singapore.
| |
Collapse
|
27
|
Bertschi NL, Voorberg-van der Wel A, Zeeman AM, Schuierer S, Nigsch F, Carbone W, Knehr J, Gupta DK, Hofman SO, van der Werff N, Nieuwenhuis I, Klooster E, Faber BW, Flannery EL, Mikolajczak SA, Chuenchob V, Shrestha B, Beibel M, Bouwmeester T, Kangwanrangsan N, Sattabongkot J, Diagana TT, Kocken CH, Roma G. Transcriptomic analysis reveals reduced transcriptional activity in the malaria parasite Plasmodium cynomolgi during progression into dormancy. eLife 2018; 7:41081. [PMID: 30589413 PMCID: PMC6344078 DOI: 10.7554/elife.41081] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 12/23/2018] [Indexed: 02/06/2023] Open
Abstract
Relapses of Plasmodium dormant liver hypnozoites compromise malaria eradication efforts. New radical cure drugs are urgently needed, yet the vast gap in knowledge of hypnozoite biology impedes drug discovery. We previously unraveled the transcriptome of 6 to 7 day-old P. cynomolgi liver stages, highlighting pathways associated with hypnozoite dormancy (Voorberg-van der Wel et al., 2017). We now extend these findings by transcriptome profiling of 9 to 10 day-old liver stage parasites, thus revealing for the first time the maturation of the dormant stage over time. Although progression of dormancy leads to a 10-fold decrease in transcription and expression of only 840 genes, including genes associated with housekeeping functions, we show that pathways involved in quiescence, energy metabolism and maintenance of genome integrity remain the prevalent pathways active in mature hypnozoites.
Collapse
Affiliation(s)
- Nicole L Bertschi
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Europe
| | | | - Anne-Marie Zeeman
- Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - Sven Schuierer
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Europe
| | - Florian Nigsch
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Europe
| | - Walter Carbone
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Europe
| | - Judith Knehr
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Europe
| | - Devendra K Gupta
- Novartis Institute for Tropical Diseases, Novartis Pharma AG, Emeryville, United States
| | - Sam O Hofman
- Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - Nicole van der Werff
- Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - Ivonne Nieuwenhuis
- Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - Els Klooster
- Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - Bart W Faber
- Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - Erika L Flannery
- Novartis Institute for Tropical Diseases, Novartis Pharma AG, Emeryville, United States
| | | | - Vorada Chuenchob
- Novartis Institute for Tropical Diseases, Novartis Pharma AG, Emeryville, United States
| | - Binesh Shrestha
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Europe
| | - Martin Beibel
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Europe
| | - Tewis Bouwmeester
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Europe
| | - Niwat Kangwanrangsan
- Department of Pathobiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Thierry T Diagana
- Novartis Institute for Tropical Diseases, Novartis Pharma AG, Emeryville, United States
| | - Clemens Hm Kocken
- Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - Guglielmo Roma
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Europe
| |
Collapse
|
28
|
Orjuela-Sanchez P, Villa ZH, Moreno M, Tong-Rios C, Meister S, LaMonte GM, Campo B, Vinetz JM, Winzeler EA. Developing Plasmodium vivax Resources for Liver Stage Study in the Peruvian Amazon Region. ACS Infect Dis 2018. [PMID: 29542317 DOI: 10.1021/acsinfecdis.7b00198] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
To develop new drugs and vaccines for malaria elimination, it will be necessary to discover biological interventions, including small molecules that act against Plasmodium vivax exoerythrocytic forms. However, a robust in vitro culture system for P. vivax is still lacking. Thus, to study exoerythrocytic forms, researchers must have simultaneous access to fresh, temperature-controlled patient blood samples, as well as an anopheline mosquito colony. In addition, researchers must rely on native mosquito species to avoid introducing a potentially dangerous invasive species into a malaria-endemic region. Here, we report an in vitro culture system carried out on site in a malaria-endemic region for liver stage parasites of P. vivax sporozoites obtained from An. darlingi, the main malaria vector in the Americas. P. vivax sporozoites were obtained by dissection of salivary glands from infected An. darlingi mosquitoes and purified by Accudenz density gradient centrifugation. HC04 liver cells were exposed to P. vivax sporozoites and cultured up to 9 days. To overcome low P. vivax patient parasitemias, potentially lower mosquito vectorial capacity, and humid, nonsterile environmental conditions, a new antibiotic cocktail was included in tissue culture to prevent contamination. Culturing conditions supported exoerythrocytic (EEF) P. vivax liver stage growth up to 9 days and allowed for maturation into intrahepatocyte merosomes. Some of the identified small forms were resistant to atovaquone (1 μM) but sensitive to the phosphatidylinositol 4-kinase inhibitor, KDU691 (1 μM). This study reports a field-accessible EEF production process for drug discovery in a malaria-endemic site in which viable P. vivax sporozoites are used for drug studies using hepatocyte infection. Our data demonstrate that the development of meaningful, field-based resources for P. vivax liver stage drug screening and liver stage human malaria experimentation in the Amazon region is feasible.
Collapse
Affiliation(s)
- Pamela Orjuela-Sanchez
- Division of Host-Microbe Systems and Therapeutics, Health Sciences Center for Immunology, Infection and Inflammation, Department of Pediatrics, School of Medicine, University of California, San Diego, 9500 Gilman Drive #0760, La Jolla, California 92093-0760, United States
| | - Zaira Hellen Villa
- Laboratorio ICEMR-Amazonia, Laboratorios de Investigacion y Desarrollo, Facultad de Ciencias y Filosofia, Universidad Peruana Cayetano Heredia, Av. Honorio Delgado 430, San Martín de Porres, Lima, 15102, Peru
| | - Marta Moreno
- Division of Infectious Diseases, Department of Medicine, University of California, San Diego, 9500 Gilman Drive #0760, La Jolla, California 92093-0760, United States
| | - Carlos Tong-Rios
- Laboratorio ICEMR-Amazonia, Laboratorios de Investigacion y Desarrollo, Facultad de Ciencias y Filosofia, Universidad Peruana Cayetano Heredia, Av. Honorio Delgado 430, San Martín de Porres, Lima, 15102, Peru
| | - Stephan Meister
- Division of Host-Microbe Systems and Therapeutics, Health Sciences Center for Immunology, Infection and Inflammation, Department of Pediatrics, School of Medicine, University of California, San Diego, 9500 Gilman Drive #0760, La Jolla, California 92093-0760, United States
| | - Gregory M. LaMonte
- Division of Host-Microbe Systems and Therapeutics, Health Sciences Center for Immunology, Infection and Inflammation, Department of Pediatrics, School of Medicine, University of California, San Diego, 9500 Gilman Drive #0760, La Jolla, California 92093-0760, United States
| | - Brice Campo
- Medicines for Malaria Venture (MMV), International Center Cointrin, Block G, 20 Route de Pre-Bois, POB 1826, Geneva, CH-1215, Switzerland
| | - Joseph M. Vinetz
- Laboratorio ICEMR-Amazonia, Laboratorios de Investigacion y Desarrollo, Facultad de Ciencias y Filosofia, Universidad Peruana Cayetano Heredia, Av. Honorio Delgado 430, San Martín de Porres, Lima, 15102, Peru
- Division of Infectious Diseases, Department of Medicine, University of California, San Diego, 9500 Gilman Drive #0760, La Jolla, California 92093-0760, United States
- Instituto de Medicina Tropical “Alexander von Humboldt”, Universidad Peruana Cayetano Heredia, Lima, 15102, Peru
| | - Elizabeth A. Winzeler
- Division of Host-Microbe Systems and Therapeutics, Health Sciences Center for Immunology, Infection and Inflammation, Department of Pediatrics, School of Medicine, University of California, San Diego, 9500 Gilman Drive #0760, La Jolla, California 92093-0760, United States
| |
Collapse
|
29
|
Voorberg-van der Wel A, Roma G, Gupta DK, Schuierer S, Nigsch F, Carbone W, Zeeman AM, Lee BH, Hofman SO, Faber BW, Knehr J, Pasini E, Kinzel B, Bifani P, Bonamy GMC, Bouwmeester T, Kocken CHM, Diagana TT. A comparative transcriptomic analysis of replicating and dormant liver stages of the relapsing malaria parasite Plasmodium cynomolgi. eLife 2017; 6:29605. [PMID: 29215331 PMCID: PMC5758109 DOI: 10.7554/elife.29605] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 12/05/2017] [Indexed: 01/23/2023] Open
Abstract
Plasmodium liver hypnozoites, which cause disease relapse, are widely considered to be the last barrier towards malaria eradication. The biology of this quiescent form of the parasite is poorly understood which hinders drug discovery. We report a comparative transcriptomic dataset of replicating liver schizonts and dormant hypnozoites of the relapsing parasite Plasmodium cynomolgi. Hypnozoites express only 34% of Plasmodium physiological pathways, while 91% are expressed in replicating schizonts. Few known malaria drug targets are expressed in quiescent parasites, but pathways involved in microbial dormancy, maintenance of genome integrity and ATP homeostasis were robustly expressed. Several transcripts encoding heavy metal transporters were expressed in hypnozoites and the copper chelator neocuproine was cidal to all liver stage parasites. This transcriptomic dataset is a valuable resource for the discovery of vaccines and effective treatments to combat vivax malaria.
Collapse
Affiliation(s)
| | - Guglielmo Roma
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | | | - Sven Schuierer
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Florian Nigsch
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Walter Carbone
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Anne-Marie Zeeman
- Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, Netherlands
| | - Boon Heng Lee
- Novartis Institute for Tropical Diseases, Singapore, Singapore
| | - Sam O Hofman
- Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, Netherlands
| | - Bart W Faber
- Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, Netherlands
| | - Judith Knehr
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Erica Pasini
- Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, Netherlands
| | - Bernd Kinzel
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Pablo Bifani
- Novartis Institute for Tropical Diseases, Singapore, Singapore
| | | | | | - Clemens H M Kocken
- Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, Netherlands
| | | |
Collapse
|
30
|
Armistead JS, Adams JH. Advancing Research Models and Technologies to Overcome Biological Barriers to Plasmodium vivax Control. Trends Parasitol 2017; 34:114-126. [PMID: 29153587 DOI: 10.1016/j.pt.2017.10.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Revised: 10/25/2017] [Accepted: 10/25/2017] [Indexed: 02/06/2023]
Abstract
Malaria prevalence has declined in the past 10 years, especially outside of sub-Saharan Africa. However, the proportion of cases due to Plasmodium vivax is increasing, accounting for up to 90-100% of the malaria burden in endemic regions. Nonetheless, investments in malaria research and control still prioritize Plasmodium falciparum while largely neglecting P. vivax. Specific biological features of P. vivax, particularly invasion of reticulocytes, occurrence of dormant liver forms of the parasite, and the potential for transmission of sexual-stage parasites prior to onset of clinical illness, promote its persistence and hinder development of research tools and interventions. This review discusses recent advances in P. vivax research, current knowledge of its unique biology, and proposes priorities for P. vivax research and control efforts.
Collapse
Affiliation(s)
- Jennifer S Armistead
- Center for Global Health and Infectious Diseases Research, Department of Global Health, College of Public Health, University of South Florida, Tampa, FL 33612, USA
| | - John H Adams
- Center for Global Health and Infectious Diseases Research, Department of Global Health, College of Public Health, University of South Florida, Tampa, FL 33612, USA.
| |
Collapse
|
31
|
Abstract
Basic science holds enormous power for revealing the biological mechanisms of disease and, in turn, paving the way toward new, effective interventions. Recognizing this power, the 2011 Research Agenda for Malaria Eradication included key priorities in fundamental research that, if attained, could help accelerate progress toward disease elimination and eradication. The Malaria Eradication Research Agenda (malERA) Consultative Panel on Basic Science and Enabling Technologies reviewed the progress, continuing challenges, and major opportunities for future research. The recommendations come from a literature of published and unpublished materials and the deliberations of the malERA Refresh Consultative Panel. These areas span multiple aspects of the Plasmodium life cycle in both the human host and the Anopheles vector and include critical, unanswered questions about parasite transmission, human infection in the liver, asexual-stage biology, and malaria persistence. We believe an integrated approach encompassing human immunology, parasitology, and entomology, and harnessing new and emerging biomedical technologies offers the best path toward addressing these questions and, ultimately, lowering the worldwide burden of malaria.
Collapse
|
32
|
Othman AS, Marin-Mogollon C, Salman AM, Franke-Fayard BM, Janse CJ, Khan SM. The use of transgenic parasites in malaria vaccine research. Expert Rev Vaccines 2017; 16:1-13. [DOI: 10.1080/14760584.2017.1333426] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Ahmad Syibli Othman
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
- Faculty of Health Sciences, Universiti Sultan Zainal Abidin, Terengganu, Malaysia
| | - Catherin Marin-Mogollon
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | | | - Blandine M. Franke-Fayard
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | - Chris J. Janse
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | - Shahid M. Khan
- Leiden Malaria Research Group, Parasitology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| |
Collapse
|
33
|
Abstract
SUMMARYThe study of malaria in the laboratory relies on either thein vitroculture of human parasites, or the use of non-human malaria parasites in laboratory animals. In this review, we address the use of non-human primate malaria parasite species (NHPMPs) in laboratory research. We describe the features of the most commonly used NHPMPs, review their contribution to our understanding of malaria to date, and discuss their potential contribution to future studies.
Collapse
|
34
|
Ang MLT, Pethe K. Contribution of high-content imaging technologies to the development of anti-infective drugs. Cytometry A 2016; 89:755-60. [PMID: 27272127 PMCID: PMC5089693 DOI: 10.1002/cyto.a.22885] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 03/17/2016] [Accepted: 05/06/2016] [Indexed: 12/18/2022]
Abstract
Originally developed to study fundamental aspects of cellular biology, high‐content imaging (HCI) was rapidly adapted to study host–pathogen interactions at the cellular level and adopted as a technology of choice to unravel disease biology. HCI platforms allow for the visualization and quantification of discrete phenotypes that cannot be captured using classical screening approaches. A key advantage of high‐content screening technologies lies in the possibility to develop and interrogate physiologically significant, predictive ex vivo disease models that reproduce complex conditions relevant for infection. Here we review and discuss recent advances in HCI technologies and chemical biology approaches that are contributing to an increased understanding of the intricate host–pathogen interrelationship on the cellular level, and which will foster the development of novel therapeutic approaches for the treatment of human bacterial and protozoan infections. © 2016 The Authors. Cytometry Part A published by Wiley Periodicals, Inc. on behalf of ISAC
Collapse
Affiliation(s)
- Michelle Lay Teng Ang
- Lee Kong Chian School of Medicine and School of Biological Sciences, Nanyang Technological University, Singapore
| | - Kevin Pethe
- Lee Kong Chian School of Medicine and School of Biological Sciences, Nanyang Technological University, Singapore
| |
Collapse
|
35
|
PI4 Kinase Is a Prophylactic but Not Radical Curative Target in Plasmodium vivax-Type Malaria Parasites. Antimicrob Agents Chemother 2016; 60:2858-63. [PMID: 26926645 PMCID: PMC4862498 DOI: 10.1128/aac.03080-15] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 02/19/2016] [Indexed: 01/04/2023] Open
Abstract
Two Plasmodium PI4 kinase (PI4K) inhibitors, KDU691 and LMV599, were selected for in vivo testing as causal prophylactic and radical-cure agents for Plasmodium cynomolgi sporozoite-infected rhesus macaques, based on their in vitro activity against liver stages. Animals were infected with P. cynomolgi sporozoites, and compounds were dosed orally. Both the KDU691 and LMV599 compounds were fully protective when administered prophylactically, and the more potent compound LMV599 achieved protection as a single oral dose of 25 mg/kg of body weight. In contrast, when tested for radical cure, five daily doses of 20 mg/kg of KDU691 or 25 mg/kg of LMV599 did not prevent relapse, as all animals experienced a secondary infection due to the reactivation of hypnozoites in the liver. Pharmacokinetic data show that LMV599 achieved plasma exposure that was sufficient to achieve efficacy based on our in vitro data. These findings indicate that Plasmodium PI4K is a potential drug target for malaria prophylaxis but not radical cure. Longer in vitro culture systems will be required to assess these compounds' activity on established hypnozoites and predict radical cure in vivo.
Collapse
|
36
|
Ang MLT, Murima P, Pethe K. Next-generation antimicrobials: from chemical biology to first-in-class drugs. Arch Pharm Res 2015; 38:1702-17. [PMID: 26259630 PMCID: PMC4567591 DOI: 10.1007/s12272-015-0645-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 07/29/2015] [Indexed: 01/11/2023]
Abstract
The global emergence of multi-drug resistant bacteria invokes an urgent and imperative necessity for the identification of novel antimicrobials. The general lack of success in progressing novel chemical entities from target-based drug screens have prompted calls for radical and innovative approaches for drug discovery. Recent developments in chemical biology and target deconvolution strategies have revived interests in the utilization of whole-cell phenotypic screens and resulted in several success stories for the discovery and development novel drug candidates and target pathways. In this review, we present and discuss recent chemical biology approaches focusing on the discovery of novel targets and new lead molecules for the treatment of human bacterial and protozoan infections.
Collapse
Affiliation(s)
- Michelle Lay Teng Ang
- Lee Kong Chian School of Medicine and School of Biological Sciences, Nanyang Technological University, 30 Biopolis Street, #B2-15a, Singapore, 138671, Singapore.
| | - Paul Murima
- Global Health Institute, Swiss Federal Institute of Technology in Lausanne (EPFL), 1015, Lausanne, Switzerland
| | - Kevin Pethe
- Lee Kong Chian School of Medicine and School of Biological Sciences, Nanyang Technological University, 30 Biopolis Street, #B2-15a, Singapore, 138671, Singapore.
| |
Collapse
|
37
|
de Koning-Ward TF, Gilson PR, Crabb BS. Advances in molecular genetic systems in malaria. Nat Rev Microbiol 2015; 13:373-87. [DOI: 10.1038/nrmicro3450] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
38
|
Campo B, Vandal O, Wesche DL, Burrows JN. Killing the hypnozoite--drug discovery approaches to prevent relapse in Plasmodium vivax. Pathog Glob Health 2015; 109:107-22. [PMID: 25891812 PMCID: PMC4455353 DOI: 10.1179/2047773215y.0000000013] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The eradication of malaria will only be possible if effective, well-tolerated medicines kill hypnozoites in vivax and ovale malaria, and thus prevent relapses in patients. Despite progress in the 8-aminoquinoline series, with tafenoquine in Phase III showing clear benefits over primaquine, the drug discovery challenge to identify hypnozoiticidal or hypnozoite-activating compounds has been hampered by the dearth of biological tools and assays, which in turn has been limited by the immense scientific and logistical challenges associated with accessing relevant human tissue and sporozoites. This review summarises the existing drug discovery series and approaches concerning the goal to block relapse.
Collapse
Affiliation(s)
- Brice Campo
- Medicines for Malaria Venture, Geneva, Switzerland
| | - Omar Vandal
- The Bill and Melinda Gates Foundation, Seattle, WA, USA
| | - David L. Wesche
- The Bill and Melinda Gates Foundation, Seattle, WA, USA
- Great Lakes Drug Development/Certara, Princeton, NJ, USA
| | | |
Collapse
|
39
|
Kumar S, Kumari R, Pandey R. New insight-guided approaches to detect, cure, prevent and eliminate malaria. PROTOPLASMA 2015; 252:717-53. [PMID: 25323622 DOI: 10.1007/s00709-014-0697-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Accepted: 09/01/2014] [Indexed: 06/04/2023]
Abstract
New challenges posed by the development of resistance against artemisinin-based combination therapies (ACTs) as well as previous first-line therapies, and the continuing absence of vaccine, have given impetus to research in all areas of malaria control. This review portrays the ongoing progress in several directions of malaria research. The variants of RTS,S and apical membrane antigen 1 (AMA1) are being developed and test adapted as multicomponent and multistage malaria control vaccines, while many other vaccine candidates and methodologies to produce antigens are under experimentation. To track and prevent the spread of artemisinin resistance from Southeast Asia to other parts of the world, rolling circle-enhanced enzyme activity detection (REEAD), a time- and cost-effective malaria diagnosis in field conditions, and a DNA marker associated with artemisinin resistance have become available. Novel mosquito repellents and mosquito trapping and killing techniques much more effective than the prevalent ones are undergoing field testing. Mosquito lines stably infected with their symbiotic wild-type or genetically engineered bacteria that kill sympatric malaria parasites are being constructed and field tested for stopping malaria transmission. A complementary approach being pursued is the addition of ivermectin-like drug molecules to ACTs to cure malaria and kill mosquitoes. Experiments are in progress to eradicate malaria mosquito by making it genetically male sterile. High-throughput screening procedures are being developed and used to discover molecules that possess long in vivo half life and are active against liver and blood stages for the fast cure of malaria symptoms caused by simple or relapsing and drug-sensitive and drug-resistant types of varied malaria parasites, can stop gametocytogenesis and sporogony and could be given in one dose. Target-based antimalarial drug designing has begun. Some of the putative next-generation antimalarials that possess in their scaffold structure several of the desired properties of malaria cure and control are exemplified by OZ439, NITD609, ELQ300 and tafenoquine that are already undergoing clinical trials, and decoquinate, usnic acid, torin-2, ferroquine, WEHI-916, MMV396749 and benzothiophene-type N-myristoyltransferase (NMT) inhibitors, which are candidates for future clinical usage. Among these, NITD609, ELQ300, decoquinate, usnic acid, torin-2 and NMT inhibitors not only cure simple malaria and are prophylactic against simple malaria, but they also cure relapsing malaria.
Collapse
Affiliation(s)
- Sushil Kumar
- SKA Institution for Research, Education and Development (SKAIRED), 4/11 SarvPriya Vihar, New Delhi, 110016, India,
| | | | | |
Collapse
|
40
|
Joyner C, Barnwell JW, Galinski MR. No more monkeying around: primate malaria model systems are key to understanding Plasmodium vivax liver-stage biology, hypnozoites, and relapses. Front Microbiol 2015; 6:145. [PMID: 25859242 PMCID: PMC4374475 DOI: 10.3389/fmicb.2015.00145] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 02/07/2015] [Indexed: 01/17/2023] Open
Abstract
Plasmodium vivax is a human malaria parasite responsible for significant morbidity worldwide and potentially death. This parasite possesses formidable liver-stage biology that involves the formation of dormant parasites known as hypnozoites. Hypnozoites are capable of activating weeks, months, or years after a primary blood-stage infection causing relapsing bouts of illness. Elimination of this dormant parasitic reservoir will be critical for global malaria eradication. Although hypnozoites were first discovered in 1982, few advancements have been made to understand their composition and biology. Until recently, in vitro models did not exist to study these forms and studying them from human ex vivo samples was virtually impossible. Today, non-human primate (NHP) models and modern systems biology approaches are poised as tools to enable the in-depth study of P. vivax liver-stage biology, including hypnozoites and relapses. NHP liver-stage model systems for P. vivax and the related simian malaria species P. cynomolgi are discussed along with perspectives regarding metabolite biomarker discovery, putative roles of extracellular vesicles, and relapse immunobiology.
Collapse
Affiliation(s)
- Chester Joyner
- Malaria Host–Pathogen Interaction Center, Emory Vaccine Center, Yerkes National Primate Research Center, Emory UniversityAtlanta, GA, USA
| | - John W. Barnwell
- Malaria Host–Pathogen Interaction Center, Emory Vaccine Center, Yerkes National Primate Research Center, Emory UniversityAtlanta, GA, USA
- Malaria Branch, Division of Parasitic Diseases and Malaria, Centers for Disease Control and PreventionAtlanta, GA, USA
| | - Mary R. Galinski
- Malaria Host–Pathogen Interaction Center, Emory Vaccine Center, Yerkes National Primate Research Center, Emory UniversityAtlanta, GA, USA
- Division of Infectious Diseases, Department of Medicine, Emory UniversityAtlanta, GA, USA
| |
Collapse
|
41
|
Plasmodium vivax liver stage development and hypnozoite persistence in human liver-chimeric mice. Cell Host Microbe 2015; 17:526-35. [PMID: 25800544 DOI: 10.1016/j.chom.2015.02.011] [Citation(s) in RCA: 160] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 07/10/2014] [Accepted: 02/16/2015] [Indexed: 12/11/2022]
Abstract
Plasmodium vivax malaria is characterized by periodic relapses of symptomatic blood stage parasite infections likely initiated by activation of dormant liver stage parasites-hypnozoites. The lack of tractable P. vivax animal models constitutes an obstacle in examining P. vivax liver stage infection and drug efficacy. To overcome this obstacle, we have used human liver-chimeric (huHep) FRG KO mice as a model for P. vivax infection. FRG KO huHep mice support P. vivax sporozoite infection, liver stage development, and hypnozoite formation. We show complete P. vivax liver stage development, including maturation into infectious exo-erythrocytic merozoites as well as the formation and persistence of hypnozoites. Prophylaxis or treatment with the antimalarial primaquine can prevent and eliminate liver stage infection, respectively. Thus, P. vivax-infected FRG KO huHep mice are a model to investigate liver stage development and dormancy and may facilitate the discovery of drugs targeting relapsing malaria.
Collapse
|
42
|
Antirelapse Efficacy of Various Primaquine Regimens for Plasmodium vivax. Malar Res Treat 2014; 2014:347018. [PMID: 25295216 PMCID: PMC4176909 DOI: 10.1155/2014/347018] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 07/28/2014] [Accepted: 08/13/2014] [Indexed: 11/18/2022] Open
Abstract
Background. Efficacy of standard dose of primaquine (PQ) as antirelapse for P. vivax has decreased. We aimed to assess efficacy of different PQ regimens. Methods. It was an open label, randomized, controlled, parallel group, assessor blind study comparing antirelapse efficacy of 3 PQ regimens (B = 15 mg/day × 14 days, C = 30 mg/day × 7 days, and D = 30 mg/day × 14 days) with no PQ group (A) in P. vivax patients. Paired primary and recurrence samples were subjected to 3 methods: (i) month of recurrence and genotyping, (ii) by PCR-RFLP, and (iii) PCR sequencing, to differentiate relapse and reinfection. The rates of recurrence relapse and reinfection were compared. Methods were compared for concordance between them. Results. The recurrence rate was 16.39%, 8.07%, 10.07%, and 6.62% in groups A, B, C, and D,
respectively (P = 0.004). The relapse rate was 6.89%, 1.55%, 4%, and 3.85% as per the month of recurrence; 8.2%, 2%, 4.58%, and 3.68% (P = 0.007) as per PCR-RFLP; and 2.73%, 1.47%, 1.55%, and 1.53% as per PCR sequencing for groups A, B, C, and D, respectively. The concordance between methods was low, 45%. Conclusion. The higher recurrence rate in no PQ as compared to PQ groups documents PQ antirelapse activity. Regimens tested were safe. However, probable resistance to PQ warrants continuous monitoring and low concordance and limitations in the methods warrant caution in interpreting.
Collapse
|
43
|
Frevert U, Nacer A. Immunobiology of Plasmodium in liver and brain. Parasite Immunol 2014; 35:267-82. [PMID: 23631610 DOI: 10.1111/pim.12039] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2013] [Accepted: 04/17/2013] [Indexed: 12/11/2022]
Abstract
Malaria remains one of the most serious health problems globally, but our understanding of the biology of the parasite and the pathogenesis of severe disease is still limited. Multiple cellular effector mechanisms that mediate parasite elimination from the liver have been described, but how effector cells use classical granule-mediated cytotoxicity to attack infected hepatocytes and how cytokines and chemokines spread via the unique fluid pathways of the liver to reach the parasites over considerable distances remains unknown. Similarly, a wealth of information on cerebral malaria (CM), one of the most severe manifestations of the disease, was gained from post-mortem analyses of human brain and murine disease models, but the cellular processes that ultimately cause disease are not fully understood. Here, we discuss how imaging of the local dynamics of parasite infection and host response as well as consideration of anatomical and physiological features of liver and brain can provide a better understanding of the initial asymptomatic hepatic phase of the infection and the cascade of events leading to CM. Given the increasing drug resistance of both parasite and vector and the unavailability of a protective vaccine, the urgency to reduce the tremendous morbidity and mortality associated with severe malaria is obvious.
Collapse
Affiliation(s)
- U Frevert
- Division of Medical Parasitology, Department of Microbiology, New York University School of Medicine, New York, NY 10010, USA.
| | | |
Collapse
|
44
|
Grüring C, Moon RW, Lim C, Holder AA, Blackman MJ, Duraisingh MT. Human red blood cell-adapted Plasmodium knowlesi parasites: a new model system for malaria research. Cell Microbiol 2014; 16:612-20. [PMID: 24506567 DOI: 10.1111/cmi.12275] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 01/24/2014] [Accepted: 01/27/2014] [Indexed: 02/06/2023]
Abstract
Plasmodium knowlesi is a simian malaria parasite primarily infecting macaque species in Southeast Asia. Although its capacity to infect humans has been recognized since the early part of the last century, it has recently become evident that human infections are widespread and potentially life threatening. Historically, P. knowlesi has proven to be a powerful tool in early studies of malaria parasites, providing key breakthroughs in understanding many aspects of Plasmodium biology. However, the necessity to grow the parasite either in macaques or in vitro using macaque blood restricted research to laboratories with access to these resources. The recent adaptation of P. knowlesi to grow and proliferate in vitro in human red blood cells (RBCs) is therefore a substantial step towards revitalizing and expanding research on P. knowlesi. Furthermore, the development of a highly efficient transfection system to genetically modify the parasite makes P. knowlesi an ideal model to study parasite biology. In this review, we elaborate on the importance of P. knowlesi in earlier phases of malaria research and highlight the future potential of the newly available human adapted P. knowlesi parasite lines.
Collapse
Affiliation(s)
- Christof Grüring
- Department of Immunology & Infectious Diseases, Harvard School of Public Health, Boston, MA, 02115, USA
| | | | | | | | | | | |
Collapse
|
45
|
Dembélé L, Franetich JF, Lorthiois A, Gego A, Zeeman AM, Kocken CHM, Le Grand R, Dereuddre-Bosquet N, van Gemert GJ, Sauerwein R, Vaillant JC, Hannoun L, Fuchter MJ, Diagana TT, Malmquist NA, Scherf A, Snounou G, Mazier D. Persistence and activation of malaria hypnozoites in long-term primary hepatocyte cultures. Nat Med 2014; 20:307-12. [PMID: 24509527 DOI: 10.1038/nm.3461] [Citation(s) in RCA: 144] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Accepted: 06/24/2013] [Indexed: 02/08/2023]
Abstract
Malaria relapses, resulting from the activation of quiescent hepatic hypnozoites of Plasmodium vivax and Plasmodium ovale, hinder global efforts to control and eliminate malaria. As primaquine, the only drug capable of eliminating hypnozoites, is unsuitable for mass administration, an alternative drug is needed urgently. Currently, analyses of hypnozoites, including screening of compounds that would eliminate them, can only be made using common macaque models, principally Macaca rhesus and Macaca fascicularis, experimentally infected with the relapsing Plasmodium cynomolgi. Here, we present a protocol for long-term in vitro cultivation of P. cynomolgi-infected M. fascicularis primary hepatocytes during which hypnozoites persist and activate to resume normal development. In a proof-of-concept experiment, we obtained evidence that exposure to an inhibitor of histone modification enzymes implicated in epigenetic control of gene expression induces an accelerated rate of hypnozoite activation. The protocol presented may further enable investigations of hypnozoite biology and the search for compounds that kill hypnozoites or disrupt their quiescence.
Collapse
Affiliation(s)
- Laurent Dembélé
- 1] Sorbonne Universités, Université Pierre et Marie Curie Paris 6, UMR S945, Paris, France. [2] Institut National de la Santé et de la Recherche Médicale, U945, Paris, France. [3]
| | - Jean-François Franetich
- 1] Sorbonne Universités, Université Pierre et Marie Curie Paris 6, UMR S945, Paris, France. [2] Institut National de la Santé et de la Recherche Médicale, U945, Paris, France. [3]
| | - Audrey Lorthiois
- 1] Sorbonne Universités, Université Pierre et Marie Curie Paris 6, UMR S945, Paris, France. [2] Institut National de la Santé et de la Recherche Médicale, U945, Paris, France
| | - Audrey Gego
- 1] Sorbonne Universités, Université Pierre et Marie Curie Paris 6, UMR S945, Paris, France. [2] Institut National de la Santé et de la Recherche Médicale, U945, Paris, France
| | - Anne-Marie Zeeman
- Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - Clemens H M Kocken
- Department of Parasitology, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - Roger Le Grand
- 1] Division of Immuno-Virology, Institute of Emerging Diseases and Innovative Therapies, Commissariat à l'Energie Atomique et aux Energies Alternatives, Fontenay-aux-Roses, France. [2] Université Paris-Sud XI, UMRE01, Orsay, France
| | - Nathalie Dereuddre-Bosquet
- 1] Division of Immuno-Virology, Institute of Emerging Diseases and Innovative Therapies, Commissariat à l'Energie Atomique et aux Energies Alternatives, Fontenay-aux-Roses, France. [2] Université Paris-Sud XI, UMRE01, Orsay, France
| | - Geert-Jan van Gemert
- Department of Medical Microbiology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Robert Sauerwein
- Department of Medical Microbiology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Jean-Christophe Vaillant
- Service de Chirurgie Digestive, Hépato-Bilio-Pancréatique et Transplantation Hépatique, Hôpital Pitié-Salpêtrière, Paris, France
| | - Laurent Hannoun
- Service de Chirurgie Digestive, Hépato-Bilio-Pancréatique et Transplantation Hépatique, Hôpital Pitié-Salpêtrière, Paris, France
| | - Matthew J Fuchter
- Department of Chemistry, Imperial College London, South Kensington Campus, London, UK
| | | | - Nicholas A Malmquist
- 1] Unité de Biologie des Interactions Hôte-Parasite, Institut Pasteur, Paris, France. [2] Centre National de la Recherche Scientifique, Unité de Recherche Associée 2581, Paris, France
| | - Artur Scherf
- 1] Unité de Biologie des Interactions Hôte-Parasite, Institut Pasteur, Paris, France. [2] Centre National de la Recherche Scientifique, Unité de Recherche Associée 2581, Paris, France
| | - Georges Snounou
- 1] Sorbonne Universités, Université Pierre et Marie Curie Paris 6, UMR S945, Paris, France. [2] Institut National de la Santé et de la Recherche Médicale, U945, Paris, France
| | - Dominique Mazier
- 1] Sorbonne Universités, Université Pierre et Marie Curie Paris 6, UMR S945, Paris, France. [2] Institut National de la Santé et de la Recherche Médicale, U945, Paris, France. [3] Assistance Publique-Hôpitaux de Paris, Groupe Hospitalier Pitié-Salpêtrière, Service Parasitologie-Mycologie, Paris, France
| |
Collapse
|
46
|
Vial H, Taramelli D, Boulton IC, Ward SA, Doerig C, Chibale K. CRIMALDDI: platform technologies and novel anti-malarial drug targets. Malar J 2013; 12:396. [PMID: 24498961 PMCID: PMC3827883 DOI: 10.1186/1475-2875-12-396] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 10/22/2013] [Indexed: 12/24/2022] Open
Abstract
The Coordination, Rationalization, and Integration of antiMALarial drug Discovery & Development Initiatives (CRIMALDDI) Consortium, funded by the EU Framework Seven Programme, has attempted, through a series of interactive and facilitated workshops, to develop priorities for research to expedite the discovery of new anti-malarials. This paper outlines the recommendations for the development of enabling technologies and the identification of novel targets.Screening systems must be robust, validated, reproducible, and represent human malaria. They also need to be cost-effective. While such systems exist to screen for activity against blood stage Plasmodium falciparum, they are lacking for other Plasmodium spp. and other stages of the parasite's life cycle. Priority needs to be given to developing high-throughput screens that can identify activity against the liver and sexual stages. This in turn requires other enabling technologies to be developed to allow the study of these stages and to allow for the culture of liver cells and the parasite at all stages of its life cycle.As these enabling technologies become available, they will allow novel drug targets to be studied. Currently anti-malarials are mostly targeting the asexual blood stage of the parasite's life cycle. There are many other attractive targets that need to be investigated. The liver stages and the sexual stages will become more important as malaria control moves towards malaria elimination. Sexual development is a process offering multiple targets, even though the mechanisms of differentiation are still not fully understood. However, designing a drug whose effect is not curative but would be used in asymptomatic patients is difficult given current safety thresholds. Compounds active against the liver schizont would have a prophylactic effect and Plasmodium vivax elimination requires effectors against the dormant liver hypnozoites. It may be that drugs to be used in elimination campaigns will also need to have utility in the control phase. Compounds with activity against blood stages need to be screened for activity against other stages.Natural products should also be a valuable source of new compounds. They often occupy non-Lipinski chemical space and so may reveal valuable new chemotypes.
Collapse
Affiliation(s)
| | | | | | - Steve A Ward
- Liverpool School of Tropical Medicine, Pembroke Place, Liverpool L3 5QA, UK.
| | | | | |
Collapse
|
47
|
Held J, Kreidenweiss A, Mordmüller B. Novel approaches in antimalarial drug discovery. Expert Opin Drug Discov 2013; 8:1325-37. [PMID: 24090219 DOI: 10.1517/17460441.2013.843522] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION The development of new antimalarial drugs remains of the utmost importance, since Plasmodium falciparum has developed resistance against nearly all chemotherapeutics in clinical use. In an effort to contain the resistance of P. falciparum against artemisinins and to further eradication efforts, studies are ongoing to identify novel and more efficacious approaches to develop antimalarials. AREAS COVERED The authors review the classical and new approaches to antimalarial drug discovery, with a special emphasis on the various stages of the parasite's life cycle and the different Plasmodium species. The authors discuss the methodologies and strategies for early efficacy testing that aim to narrow down the portfolio of promising compounds. EXPERT OPINION The increased efforts in the discovery and development of new antimalarial compounds have led to the recognition of new promising hits. However, there is still major roadblock of selecting the most promising compounds and then further testing them in early clinical trials, especially in the current restricted economy. Controlled human malaria infection has much potential for speeding-up the early development process of many drug candidates including those which target the pre-erythrocytic stages.
Collapse
Affiliation(s)
- Jana Held
- University of Tübingen, Institute of Tropical Medicine , Wilhelmstraße 27, D-72074 Tübingen , Germany +49 7071 29 82364 ; +49 7071 295189 ;
| | | | | |
Collapse
|
48
|
In vivo imaging in NHP models of malaria: challenges, progress and outlooks. Parasitol Int 2013; 63:206-15. [PMID: 24042056 PMCID: PMC7108422 DOI: 10.1016/j.parint.2013.09.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Revised: 08/30/2013] [Accepted: 09/06/2013] [Indexed: 12/22/2022]
Abstract
Animal models of malaria, mainly mice, have made a large contribution to our knowledge of host-pathogen interactions and immune responses, and to drug and vaccine design. Non-human primate (NHP) models for malaria are admittedly under-used, although they are probably closer models than mice for human malaria; in particular, NHP models allow the use of human pathogens (Plasmodium falciparum, Plasmodium vivax, Plasmodium malariae and Plasmodium knowlesi). NHPs, whether natural hosts or experimentally challenged with a simian Plasmodium, can also serve as robust pre-clinical models. Some simian parasites are closely related to a human counterpart, with which they may share a common ancestor, and display similar major features with the human infection and pathology. NHP models allow longitudinal studies, from the early events following sporozoite inoculation to the later events, including analysis of organs and tissues, particularly liver, spleen, brain and bone marrow. NHP models have one other significant advantage over mouse models: NHPs are our closest relatives and thus their biology is very similar to ours. Recently developed in vivo imaging tools have provided insight into malaria parasite infection and disease in mouse models. One advantage of these tools is that they limit the need for invasive procedures, such as tissue biopsies. Many such technologies are now available for NHP studies and provide new opportunities for elucidating host/parasite interactions. The aim of this review is to bring the malaria community up to date on what is currently possible and what soon will be, in terms of in vivo imaging in NHP models of malaria, to consider the pros and the cons of the various techniques, and to identify challenges.
Collapse
|