1
|
Loveridge KM, Sigala PA. Identification of a divalent metal transporter required for cellular iron metabolism in malaria parasites. Proc Natl Acad Sci U S A 2024; 121:e2411631121. [PMID: 39467134 DOI: 10.1073/pnas.2411631121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 09/23/2024] [Indexed: 10/30/2024] Open
Abstract
Plasmodium falciparum malaria parasites invade and multiply inside red blood cells (RBCs), the most iron-rich compartment in humans. Like all cells, P. falciparum requires nutritional iron to support essential metabolic pathways, but the critical mechanisms of iron acquisition and trafficking during RBC infection have remained obscure. Parasites internalize and liberate massive amounts of heme during large-scale digestion of RBC hemoglobin within an acidic food vacuole (FV) but lack a heme oxygenase to release porphyrin-bound iron. Although most FV heme is sequestered into inert hemozoin crystals, prior studies indicate that trace heme escapes biomineralization and is susceptible to nonenzymatic degradation within the oxidizing FV environment to release labile iron. Parasites retain a homolog of divalent metal transporter 1 (DMT1), a known mammalian iron transporter, but its role in P. falciparum iron acquisition has not been tested. Our phylogenetic studies indicate that P. falciparum DMT1 (PfDMT1) retains conserved molecular features critical for metal transport. We localized this protein to the FV membrane and defined its orientation in an export-competent topology. Conditional knockdown of PfDMT1 expression is lethal to parasites, which display broad cellular defects in iron-dependent functions, including impaired apicoplast biogenesis and mitochondrial polarization. Parasites are selectively rescued from partial PfDMT1 knockdown by supplementation with exogenous iron, but not other metals. These results support a cellular paradigm whereby PfDMT1 is the molecular gatekeeper to essential iron acquisition by blood-stage malaria parasites and suggest that therapeutic targeting of PfDMT1 may be a potent antimalarial strategy.
Collapse
Affiliation(s)
- Kade M Loveridge
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84112
| | - Paul A Sigala
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT 84112
| |
Collapse
|
2
|
Andrade CM, Carrasquilla M, Dabbas U, Briggs J, van Dijk H, Sergeev N, Sissoko A, Niangaly M, Ntalla C, LaVerriere E, Skinner J, Golob K, Richter J, Cisse H, Li S, Hendry JA, Asghar M, Doumtabe D, Farnert A, Ruppert T, Neafsey DE, Kayentao K, Doumbo S, Ongoiba A, Crompton PD, Traore B, Greenhouse B, Portugal S. Infection length and host environment influence on Plasmodium falciparum dry season reservoir. EMBO Mol Med 2024; 16:2349-2375. [PMID: 39284949 PMCID: PMC11473648 DOI: 10.1038/s44321-024-00127-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: 07/27/2023] [Revised: 08/07/2024] [Accepted: 08/09/2024] [Indexed: 10/16/2024] Open
Abstract
Persistence of malaria parasites in asymptomatic hosts is crucial in areas of seasonally-interrupted transmission, where P. falciparum bridges wet seasons months apart. During the dry season, infected erythrocytes exhibit extended circulation with reduced cytoadherence, increasing the risk of splenic clearance of infected cells and hindering parasitaemia increase. However, what determines parasite persistence for long periods of time remains unknown. Here, we investigated whether seasonality affects plasma composition so that P. falciparum can detect and adjust to changing serological cues; or if alternatively, parasite infection length dictates clinical presentation and persistency. Data from Malian children exposed to alternating ~6-month wet and dry seasons show that plasma composition is unrelated to time of year in non-infected children, and that carrying P. falciparum only minimally affects plasma constitution in asymptomatic hosts. Parasites persisting in the blood of asymptomatic children from the dry into the ensuing wet season rarely if ever appeared to cause malaria in their hosts as seasons changed. In vitro culture in the presence of plasma collected in the dry or the wet seasons did not affect parasite development, replication or host-cell remodelling. The absence of a parasite-encoded sensing mechanism was further supported by the observation of similar features in P. falciparum persisting asymptomatically in the dry season and parasites in age- and sex-matched asymptomatic children in the wet season. Conversely, we show that P. falciparum clones transmitted early in the wet season had lower chance of surviving until the end of the following dry season, contrasting with a higher likelihood of survival of clones transmitted towards the end of the wet season, allowing for the re-initiation of transmission. We propose that the decreased virulence observed in persisting parasites during the dry season is not due to the parasites sensing ability, nor is it linked to a decreased capacity for parasite replication but rather a consequence decreased cytoadhesion associated with infection length.
Collapse
Affiliation(s)
- Carolina M Andrade
- Center for Infectious Diseases, Parasitology, Heidelberg University Hospital, Heidelberg, Germany.
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands.
| | | | - Usama Dabbas
- Center for Infectious Diseases, Parasitology, Heidelberg University Hospital, Heidelberg, Germany
- Max Planck Institute for Infection Biology, Berlin, Germany
| | - Jessica Briggs
- Department of Medicine, San Francisco General Hospital, University of California, San Francisco, CA, USA
- Chan-Zuckerberg Biohub, San Francisco, CA, USA
| | - Hannah van Dijk
- Center for Infectious Diseases, Parasitology, Heidelberg University Hospital, Heidelberg, Germany
| | - Nikolay Sergeev
- Center for Infectious Diseases, Parasitology, Heidelberg University Hospital, Heidelberg, Germany
| | - Awa Sissoko
- Mali International Center of Excellence in Research, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Moussa Niangaly
- Mali International Center of Excellence in Research, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | | | - Emily LaVerriere
- Department of Immunology and Infectious Diseases, Harvard TH Chan School of Public Health, Boston, MA, USA
- Broad Institute, Cambridge, MA, USA
| | - Jeff Skinner
- Laboratory of Immunogenetics, NIAID, National Institutes of Health, Rockville, MD, USA
| | - Klara Golob
- Max Planck Institute for Infection Biology, Berlin, Germany
| | - Jeremy Richter
- Max Planck Institute for Infection Biology, Berlin, Germany
| | - Hamidou Cisse
- Mali International Center of Excellence in Research, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Shanping Li
- Laboratory of Immunogenetics, NIAID, National Institutes of Health, Rockville, MD, USA
| | - Jason A Hendry
- Max Planck Institute for Infection Biology, Berlin, Germany
| | - Muhammad Asghar
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institute, Stockholm Sweden and Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
- Department of Biology, Lund University, Lund, Sweden
- Department of Healthcare Biotechnology, Atta-Ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Didier Doumtabe
- Mali International Center of Excellence in Research, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Anna Farnert
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institute, Stockholm Sweden and Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | | | - Daniel E Neafsey
- Department of Immunology and Infectious Diseases, Harvard TH Chan School of Public Health, Boston, MA, USA
- Broad Institute, Cambridge, MA, USA
| | - Kassoum Kayentao
- Mali International Center of Excellence in Research, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Safiatou Doumbo
- Mali International Center of Excellence in Research, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Aissata Ongoiba
- Mali International Center of Excellence in Research, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Peter D Crompton
- Laboratory of Immunogenetics, NIAID, National Institutes of Health, Rockville, MD, USA
| | - Boubacar Traore
- Mali International Center of Excellence in Research, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Bryan Greenhouse
- Department of Medicine, San Francisco General Hospital, University of California, San Francisco, CA, USA
- Chan-Zuckerberg Biohub, San Francisco, CA, USA
| | - Silvia Portugal
- Center for Infectious Diseases, Parasitology, Heidelberg University Hospital, Heidelberg, Germany.
- Max Planck Institute for Infection Biology, Berlin, Germany.
| |
Collapse
|
3
|
Chahine Z, Abel S, Hollin T, Barnes GL, Chung JH, Daub ME, Renard I, Choi JY, Vydyam P, Pal A, Alba-Argomaniz M, Banks CAS, Kirkwood J, Saraf A, Camino I, Castaneda P, Cuevas MC, De Mercado-Arnanz J, Fernandez-Alvaro E, Garcia-Perez A, Ibarz N, Viera-Morilla S, Prudhomme J, Joyner CJ, Bei AK, Florens L, Ben Mamoun C, Vanderwal CD, Le Roch KG. A kalihinol analog disrupts apicoplast function and vesicular trafficking in P. falciparum malaria. Science 2024; 385:eadm7966. [PMID: 39325875 DOI: 10.1126/science.adm7966] [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: 11/07/2023] [Revised: 03/27/2024] [Accepted: 07/09/2024] [Indexed: 09/28/2024]
Abstract
We report the discovery of MED6-189, an analog of the kalihinol family of isocyanoterpene natural products that is effective against drug-sensitive and drug-resistant Plasmodium falciparum strains, blocking both asexual replication and sexual differentiation. In vivo studies using a humanized mouse model of malaria confirm strong efficacy of the compound in animals with no apparent hemolytic activity or toxicity. Complementary chemical, molecular, and genomics analyses revealed that MED6-189 targets the parasite apicoplast and acts by inhibiting lipid biogenesis and cellular trafficking. Genetic analyses revealed that a mutation in PfSec13, which encodes a component of the parasite secretory machinery, reduced susceptibility to the drug. Its high potency, excellent therapeutic profile, and distinctive mode of action make MED6-189 an excellent addition to the antimalarial drug pipeline.
Collapse
Affiliation(s)
- Z Chahine
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, CA, USA
| | - S Abel
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, CA, USA
| | - T Hollin
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, CA, USA
| | - G L Barnes
- Department of Chemistry, University of California, Irvine, CA, USA
| | - J H Chung
- Department of Chemistry, University of California, Irvine, CA, USA
| | - M E Daub
- Department of Chemistry, University of California, Irvine, CA, USA
| | - I Renard
- Department of Internal Medicine, Section of Infectious Diseases, Yale School of Medicine, New Haven, CT, USA
| | - J Y Choi
- Department of Internal Medicine, Section of Infectious Diseases, Yale School of Medicine, New Haven, CT, USA
| | - P Vydyam
- Department of Internal Medicine, Section of Infectious Diseases, Yale School of Medicine, New Haven, CT, USA
| | - A Pal
- Department of Internal Medicine, Section of Infectious Diseases, Yale School of Medicine, New Haven, CT, USA
| | - M Alba-Argomaniz
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, USA
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
| | - C A S Banks
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - J Kirkwood
- Metabolomics Core Facility, University of California, Riverside, CA, USA
| | - A Saraf
- Stowers Institute for Medical Research, Kansas City, MO, USA
- Present address: Shankel Structural Biology Center, The University of Kansas, Lawrence, KS, USA
| | - I Camino
- GSK, Tres Cantos (Madrid), Spain
| | | | | | | | | | | | - N Ibarz
- GSK, Tres Cantos (Madrid), Spain
| | | | - J Prudhomme
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, CA, USA
| | - C J Joyner
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, USA
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
| | - A K Bei
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, CT, USA
| | - L Florens
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - C Ben Mamoun
- Department of Internal Medicine, Section of Infectious Diseases, Yale School of Medicine, New Haven, CT, USA
| | - C D Vanderwal
- Department of Chemistry, University of California, Irvine, CA, USA
- Department of Pharmaceutical Sciences, University of California, Irvine, CA, USA
| | - K G Le Roch
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, CA, USA
| |
Collapse
|
4
|
Loveridge KM, Sigala PA. Identification of a divalent metal transporter required for cellular iron metabolism in malaria parasites. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.10.587216. [PMID: 38798484 PMCID: PMC11118319 DOI: 10.1101/2024.05.10.587216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Plasmodium falciparum malaria parasites invade and multiply inside red blood cells (RBCs), the most iron-rich compartment in humans. Like all cells, P. falciparum requires nutritional iron to support essential metabolic pathways, but the critical mechanisms of iron acquisition and trafficking during RBC infection have remained obscure. Parasites internalize and liberate massive amounts of heme during large-scale digestion of RBC hemoglobin within an acidic food vacuole (FV) but lack a heme oxygenase to release porphyrin-bound iron. Although most FV heme is sequestered into inert hemozoin crystals, prior studies indicate that trace heme escapes biomineralization and is susceptible to non-enzymatic degradation within the oxidizing FV environment to release labile iron. Parasites retain a homolog of divalent metal transporter 1 (DMT1), a known mammalian iron transporter, but its role in P. falciparum iron acquisition has not been tested. Our phylogenetic studies indicate that P. falciparum DMT1 (PfDMT1) retains conserved molecular features critical for metal transport. We localized this protein to the FV membrane and defined its orientation in an export-competent topology. Conditional knockdown of PfDMT1 expression is lethal to parasites, which display broad cellular defects in iron-dependent functions, including impaired apicoplast biogenesis and mitochondrial polarization. Parasites are selectively rescued from partial PfDMT1 knockdown by supplementation with exogenous iron, but not other metals. These results support a cellular paradigm whereby PfDMT1 is the molecular gatekeeper to essential iron acquisition by blood-stage malaria parasites and suggest that therapeutic targeting of PfDMT1 may be a potent antimalarial strategy.
Collapse
Affiliation(s)
- Kade M. Loveridge
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Paul A. Sigala
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, United States
| |
Collapse
|
5
|
Christensen P, Cinzah R, Suwanarusk R, Chua ACY, Kaneko O, Kyle DE, Aung HL, Matheson J, Bifani P, Rénia L, Cook GM, Snounou G, Russell B. Extended blood stage sensitivity profiles of Plasmodium cynomolgi to doxycycline and tafenoquine, as a model for Plasmodium vivax. Antimicrob Agents Chemother 2024; 68:e0028024. [PMID: 38587391 PMCID: PMC11064600 DOI: 10.1128/aac.00280-24] [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: 02/22/2024] [Accepted: 03/15/2024] [Indexed: 04/09/2024] Open
Abstract
Testing Plasmodium vivax antimicrobial sensitivity is limited to ex vivo schizont maturation assays, which preclude determining the IC50s of delayed action antimalarials such as doxycycline. Using Plasmodium cynomolgi as a model for P. vivax, we determined the physiologically significant delayed death effect induced by doxycycline [IC50(96 h), 1,401 ± 607 nM]. As expected, IC50(96 h) to chloroquine (20.4 nM), piperaquine (12.6 µM), and tafenoquine (1,424 nM) were not affected by extended exposure.
Collapse
Affiliation(s)
- Peter Christensen
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Rosy Cinzah
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Rossarin Suwanarusk
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Adeline Chiew Yen Chua
- A*STAR Infectious Diseases Labs, Agency for Science, Technology and Research (A*STAR), , Singapore
| | - Osamu Kaneko
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Sakamoto, Nagasaki, Japan
| | - Dennis E. Kyle
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia, USA
| | - Htin Lin Aung
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Jessica Matheson
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Pablo Bifani
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Laurent Rénia
- A*STAR Infectious Diseases Labs, Agency for Science, Technology and Research (A*STAR), , Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Gregory M. Cook
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Georges Snounou
- 11-Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184), Fontenay-aux-Roses & Kremlin- Bicêtre, Le Kremlin-Bicêtre, France
| | - Bruce Russell
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Sakamoto, Nagasaki, Japan
| |
Collapse
|
6
|
van Schalkwyk DA, Pratt S, Nolder D, Stewart LB, Liddy H, Muwanguzi-Karugaba J, Beshir KB, Britten D, Victory E, Rogers C, Millard J, Brown M, Nabarro LE, Taylor A, Young BC, Chiodini PL, Sutherland CJ. Treatment Failure in a UK Malaria Patient Harboring Genetically Variant Plasmodium falciparum From Uganda With Reduced In Vitro Susceptibility to Artemisinin and Lumefantrine. Clin Infect Dis 2024; 78:445-452. [PMID: 38019958 PMCID: PMC10874266 DOI: 10.1093/cid/ciad724] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 11/20/2023] [Accepted: 11/27/2023] [Indexed: 12/01/2023] Open
Abstract
BACKGROUND Recent cases of clinical failure in malaria patients in the United Kingdom (UK) treated with artemether-lumefantrine have implications for malaria chemotherapy worldwide. METHODS Parasites were isolated from an index case of confirmed Plasmodium falciparum treatment failure after standard treatment, and from comparable travel-acquired UK malaria cases. Drug susceptibility in vitro and genotypes at 6 resistance-associated loci were determined for all parasite isolates and compared with clinical outcomes for each parasite donor. RESULTS A traveler, who returned to the UK from Uganda in 2022 with Plasmodium falciparum malaria, twice failed treatment with full courses of artemether-lumefantrine. Parasites from the patient exhibited significantly reduced susceptibility to artemisinin (ring-stage survival, 17.3% [95% confidence interval {CI}, 13.6%-21.1%]; P < .0001) and lumefantrine (effective concentration preventing 50% of growth = 259.4 nM [95% CI, 130.6-388.2 nM]; P = .001). Parasite genotyping identified an allele of pfk13 encoding both the A675V variant in the Pfk13 propeller domain and a novel L145V nonpropeller variant. In vitro susceptibility testing of 6 other P. falciparum lines of Ugandan origin identified reduced susceptibility to artemisinin and lumefantrine in 1 additional line, also from a 2022 treatment failure case. These parasites did not harbor a pfk13 propeller domain variant but rather the novel nonpropeller variant T349I. Variant alleles of pfubp1, pfap2mu, and pfcoronin were also identified among the 7 parasite lines. CONCLUSIONS We confirm, in a documented case of artemether-lumefantrine treatment failure imported from Uganda, the presence of pfk13 mutations encoding L145V and A675V. Parasites with reduced susceptibility to both artemisinin and lumefantrine may be emerging in Uganda.
Collapse
Affiliation(s)
- Donelly A van Schalkwyk
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Sade Pratt
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Debbie Nolder
- UK Health Security Agency Malaria Reference Laboratory, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Lindsay B Stewart
- UK Health Security Agency Malaria Reference Laboratory, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Helen Liddy
- UK Health Security Agency Malaria Reference Laboratory, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Julian Muwanguzi-Karugaba
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Khalid B Beshir
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Dawn Britten
- UK Health Security Agency Malaria Reference Laboratory, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Emma Victory
- UK Health Security Agency Malaria Reference Laboratory, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Claire Rogers
- UK Health Security Agency Malaria Reference Laboratory, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - James Millard
- Hospital for Tropical Diseases, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Michael Brown
- Hospital for Tropical Diseases, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Laura E Nabarro
- Hospital for Tropical Diseases, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Andrew Taylor
- Department of Infectious Diseases, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Bernadette C Young
- Department of Infectious Diseases, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Peter L Chiodini
- UK Health Security Agency Malaria Reference Laboratory, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Colin J Sutherland
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
- UK Health Security Agency Malaria Reference Laboratory, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
7
|
Kobpornchai P, Imwong M, Kulkeaw K. Trio fluorophore-based phenotypic assay for the detection of artemisinin-induced growth-arrested Plasmodium falciparum in human erythrocytes. Sci Rep 2024; 14:1802. [PMID: 38245618 PMCID: PMC10799909 DOI: 10.1038/s41598-024-52414-8] [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: 07/14/2023] [Accepted: 01/18/2024] [Indexed: 01/22/2024] Open
Abstract
Artemisinin combination therapy remains effective for the treatment of falciparum malaria. However, Plasmodium falciparum can escape the effects of artemisinin by arresting their growth. The growth-arrested parasites cannot be distinguished from nonviable parasites with standard microscopy techniques due to their morphological similarities. Here, we demonstrated the efficacy of a new laboratory assay that is compatible with the artemisinin susceptibility test. As a result of the differential cell permeabilities of two DNA-binding fluorophores, growth-arrested P. falciparum can be distinguished from parasites killed by artemisinin, since the latter lose cell membrane permeability. This fluorescence-based assay increased the sensitivity and specificity of the ring survival assay in the assessment of artemisinin susceptibility. When combined with a third fluorophore-conjugated anti-human leukocyte antibody, this trio fluorophore assay became more useful in identifying growth-arrested parasites in mock human blood samples. This novel assay is a simple and rapid technique for monitoring artemisinin resistance with greater sensitivity and accuracy compared with morphology-based observations under a light microscope.
Collapse
Affiliation(s)
- Porntida Kobpornchai
- Siriraj Integrative Center for Neglected Parasitic Diseases, Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
- Siriraj-Long Read Lab, Department of Bioinformatics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Mallika Imwong
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10700, Thailand
| | - Kasem Kulkeaw
- Siriraj Integrative Center for Neglected Parasitic Diseases, Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand.
| |
Collapse
|
8
|
Chahine Z, Abel S, Hollin T, Chung JH, Barnes GL, Daub ME, Renard I, Choi JY, Pratap V, Pal A, Alba-Argomaniz M, Banks CAS, Kirkwood J, Saraf A, Camino I, Castaneda P, Cuevas MC, De Mercado-Arnanz J, Fernandez-Alvaro E, Garcia-Perez A, Ibarz N, Viera-Morilla S, Prudhomme J, Joyner CJ, Bei AK, Florens L, Ben Mamoun C, Vanderwal CD, Le Roch KG. A Potent Kalihinol Analogue Disrupts Apicoplast Function and Vesicular Trafficking in P. falciparum Malaria. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.21.568162. [PMID: 38045341 PMCID: PMC10690269 DOI: 10.1101/2023.11.21.568162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Here we report the discovery of MED6-189, a new analogue of the kalihinol family of isocyanoterpene (ICT) natural products. MED6-189 is effective against drug-sensitive and -resistant P. falciparum strains blocking both intraerythrocytic asexual replication and sexual differentiation. This compound was also effective against P. knowlesi and P. cynomolgi. In vivo efficacy studies using a humanized mouse model of malaria confirms strong efficacy of the compound in animals with no apparent hemolytic activity or apparent toxicity. Complementary chemical biology, molecular biology, genomics and cell biological analyses revealed that MED6-189 primarily targets the parasite apicoplast and acts by inhibiting lipid biogenesis and cellular trafficking. Genetic analyses in P. falciparum revealed that a mutation in PfSec13, which encodes a component of the parasite secretory machinery, reduced susceptibility to the drug. The high potency of MED6-189 in vitro and in vivo, its broad range of efficacy, excellent therapeutic profile, and unique mode of action make it an excellent addition to the antimalarial drug pipeline.
Collapse
Affiliation(s)
- Z Chahine
- Department of Molecular, Cell and Systems Biology, University of California Riverside, CA, USA
| | - S Abel
- Department of Molecular, Cell and Systems Biology, University of California Riverside, CA, USA
| | - T Hollin
- Department of Molecular, Cell and Systems Biology, University of California Riverside, CA, USA
| | - JH Chung
- Department of Chemistry, University of California, Irvine, California, 92617, USA
| | - GL Barnes
- Department of Chemistry, University of California, Irvine, California, 92617, USA
| | - ME Daub
- Department of Chemistry, University of California, Irvine, California, 92617, USA
| | - I Renard
- Department of Internal Medicine, Section of Infectious Diseases, Yale School of Medicine, New Haven, Connecticut, USA
| | - JY Choi
- Department of Internal Medicine, Section of Infectious Diseases, Yale School of Medicine, New Haven, Connecticut, USA
| | - V Pratap
- Department of Internal Medicine, Section of Infectious Diseases, Yale School of Medicine, New Haven, Connecticut, USA
| | - A Pal
- Department of Internal Medicine, Section of Infectious Diseases, Yale School of Medicine, New Haven, Connecticut, USA
| | - M Alba-Argomaniz
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, United States
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, United States
| | - CAS Banks
- Stowers Institute for Medical Research, 1000 E. 50th Street, Kansas City, MO 64110, USA
| | - J Kirkwood
- Metabolomics Core Facility, University of California, Riverside, CA 92521, USA
| | - A Saraf
- Stowers Institute for Medical Research, 1000 E. 50th Street, Kansas City, MO 64110, USA
| | - I Camino
- GSK, C/ Severo Ochoa, 2 PTM, 28760 Tres Cantos (Madrid), Spain
| | - P Castaneda
- GSK, C/ Severo Ochoa, 2 PTM, 28760 Tres Cantos (Madrid), Spain
| | - MC Cuevas
- GSK, C/ Severo Ochoa, 2 PTM, 28760 Tres Cantos (Madrid), Spain
| | | | | | - A Garcia-Perez
- GSK, C/ Severo Ochoa, 2 PTM, 28760 Tres Cantos (Madrid), Spain
| | - N Ibarz
- GSK, C/ Severo Ochoa, 2 PTM, 28760 Tres Cantos (Madrid), Spain
| | - S Viera-Morilla
- GSK, C/ Severo Ochoa, 2 PTM, 28760 Tres Cantos (Madrid), Spain
| | - J Prudhomme
- Department of Molecular, Cell and Systems Biology, University of California Riverside, CA, USA
| | - CJ Joyner
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, United States
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, United States
| | - AK Bei
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, USA
| | - L Florens
- Stowers Institute for Medical Research, 1000 E. 50th Street, Kansas City, MO 64110, USA
| | - C Ben Mamoun
- Department of Internal Medicine, Section of Infectious Diseases, Yale School of Medicine, New Haven, Connecticut, USA
| | - CD Vanderwal
- Department of Chemistry, University of California, Irvine, California, 92617, USA
| | - KG Le Roch
- Department of Molecular, Cell and Systems Biology, University of California Riverside, CA, USA
| |
Collapse
|
9
|
Mok S, Yeo T, Hong D, Shears MJ, Ross LS, Ward KE, Dhingra SK, Kanai M, Bridgford JL, Tripathi AK, Mlambo G, Burkhard AY, Ansbro MR, Fairhurst KJ, Gil-Iturbe E, Park H, Rozenberg FD, Kim J, Mancia F, Fairhurst RM, Quick M, Uhlemann AC, Sinnis P, Fidock DA. Mapping the genomic landscape of multidrug resistance in Plasmodium falciparum and its impact on parasite fitness. SCIENCE ADVANCES 2023; 9:eadi2364. [PMID: 37939186 PMCID: PMC10631731 DOI: 10.1126/sciadv.adi2364] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 10/06/2023] [Indexed: 11/10/2023]
Abstract
Drug-resistant Plasmodium falciparum parasites have swept across Southeast Asia and now threaten Africa. By implementing a P. falciparum genetic cross using humanized mice, we report the identification of key determinants of resistance to artemisinin (ART) and piperaquine (PPQ) in the dominant Asian KEL1/PLA1 lineage. We mapped k13 as the central mediator of ART resistance in vitro and identified secondary markers. Applying bulk segregant analysis, quantitative trait loci mapping using 34 recombinant haplotypes, and gene editing, our data reveal an epistatic interaction between mutant PfCRT and multicopy plasmepsins 2/3 in mediating high-grade PPQ resistance. Susceptibility and parasite fitness assays implicate PPQ as a driver of selection for KEL1/PLA1 parasites. Mutant PfCRT enhanced susceptibility to lumefantrine, the first-line partner drug in Africa, highlighting a potential benefit of opposing selective pressures with this drug and PPQ. We also identified that the ABCI3 transporter can operate in concert with PfCRT and plasmepsins 2/3 in mediating multigenic resistance to antimalarial agents.
Collapse
Affiliation(s)
- Sachel Mok
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, NY, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Tomas Yeo
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, NY, USA
| | - Davin Hong
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, NY, USA
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Melanie J. Shears
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Leila S. Ross
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Kurt E. Ward
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, NY, USA
| | - Satish K. Dhingra
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Mariko Kanai
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, NY, USA
| | - Jessica L. Bridgford
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, NY, USA
| | - Abhai K. Tripathi
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Godfree Mlambo
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Anna Y. Burkhard
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Megan R. Ansbro
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Kate J. Fairhurst
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, NY, USA
| | - Eva Gil-Iturbe
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Heekuk Park
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Felix D. Rozenberg
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Jonathan Kim
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY, USA
| | - Filippo Mancia
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY, USA
| | - Rick M. Fairhurst
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Matthias Quick
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY, USA
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, USA
| | - Anne-Catrin Uhlemann
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, NY, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Photini Sinnis
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - David A. Fidock
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, NY, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
10
|
Mok S, Yeo T, Hong D, Shears MJ, Ross LS, Ward KE, Dhingra SK, Kanai M, Bridgford JL, Tripathi AK, Mlambo G, Burkhard AY, Fairhurst KJ, Gil-Iturbe E, Park H, Rozenberg FD, Kim J, Mancia F, Quick M, Uhlemann AC, Sinnis P, Fidock DA. Mapping the genomic landscape of multidrug resistance in Plasmodium falciparum and its impact on parasite fitness. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.02.543338. [PMID: 37398288 PMCID: PMC10312498 DOI: 10.1101/2023.06.02.543338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Drug-resistant Plasmodium falciparum parasites have swept across Southeast Asia and now threaten Africa. By implementing a P. falciparum genetic cross using humanized mice, we report the identification of key determinants of resistance to artemisinin (ART) and piperaquine (PPQ) in the dominant Asian KEL1/PLA1 lineage. We mapped k13 as the central mediator of ART resistance and identified secondary markers. Applying bulk segregant analysis, quantitative trait loci mapping and gene editing, our data reveal an epistatic interaction between mutant PfCRT and multicopy plasmepsins 2/3 in mediating high-grade PPQ resistance. Susceptibility and parasite fitness assays implicate PPQ as a driver of selection for KEL1/PLA1 parasites. Mutant PfCRT enhanced susceptibility to lumefantrine, the first-line partner drug in Africa, highlighting a potential benefit of opposing selective pressures with this drug and PPQ. We also identified that the ABCI3 transporter can operate in concert with PfCRT and plasmepsins 2/3 in mediating multigenic resistance to antimalarial agents.
Collapse
Affiliation(s)
- Sachel Mok
- Department of Microbiology & Immunology, Columbia University Irving Medical Center, New York, NY
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, NY
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Tomas Yeo
- Department of Microbiology & Immunology, Columbia University Irving Medical Center, New York, NY
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, NY
| | - Davin Hong
- Department of Microbiology & Immunology, Columbia University Irving Medical Center, New York, NY
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, NY
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Melanie J Shears
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Leila S Ross
- Department of Microbiology & Immunology, Columbia University Irving Medical Center, New York, NY
| | - Kurt E Ward
- Department of Microbiology & Immunology, Columbia University Irving Medical Center, New York, NY
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, NY
| | - Satish K Dhingra
- Department of Microbiology & Immunology, Columbia University Irving Medical Center, New York, NY
| | - Mariko Kanai
- Department of Microbiology & Immunology, Columbia University Irving Medical Center, New York, NY
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, NY
| | - Jessica L Bridgford
- Department of Microbiology & Immunology, Columbia University Irving Medical Center, New York, NY
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, NY
| | - Abhai K Tripathi
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Godfree Mlambo
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Anna Y Burkhard
- Department of Microbiology & Immunology, Columbia University Irving Medical Center, New York, NY
| | - Kate J Fairhurst
- Department of Microbiology & Immunology, Columbia University Irving Medical Center, New York, NY
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, NY
| | - Eva Gil-Iturbe
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Heekuk Park
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Felix D Rozenberg
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Jonathan Kim
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY, USA
| | - Filippo Mancia
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY, USA
| | - Matthias Quick
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, NY, USA
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, USA
| | - Anne-Catrin Uhlemann
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, NY
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| | - Photini Sinnis
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - David A Fidock
- Department of Microbiology & Immunology, Columbia University Irving Medical Center, New York, NY
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, NY
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY
| |
Collapse
|
11
|
Azmi WA, Rizki AFM, Djuardi Y, Artika IM, Siregar JE. Molecular insights into artemisinin resistance in Plasmodium falciparum: An updated review. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2023:105460. [PMID: 37269964 DOI: 10.1016/j.meegid.2023.105460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/25/2023] [Accepted: 05/27/2023] [Indexed: 06/05/2023]
Abstract
Malaria still poses a major burden on human health around the world, especially in endemic areas. Plasmodium resistance to several antimalarial drugs has been one of the major hindrances in control of malaria. Thus, the World Health Organization recommended artemisinin-based combination therapy (ACT) as a front-line treatment for malaria. The emergence of parasites resistant to artemisinin, along with resistant to ACT partner drugs, has led to ACT treatment failure. The artemisinin resistance is mostly related to the mutations in the propeller domain of the kelch13 (k13) gene that encodes protein Kelch13 (K13). The K13 protein has an important role in parasite reaction to oxidative stress. The most widely spread mutation in K13, with the highest degree of resistance, is a C580Y mutation. Other mutations, which are already identified as markers of artemisinin resistance, are R539T, I543T, and Y493H. The objective of this review is to provide current molecular insights into artemisinin resistance in Plasmodium falciparum. The trending use of artemisinin beyond its antimalarial effect is described. Immediate challenges and future research directions are discussed. Better understanding of the molecular mechanisms underlying artemisinin resistance will accelerate implementation of scientific findings to solve problems with malarial infection.
Collapse
Affiliation(s)
- Wihda Aisarul Azmi
- Eijkman Research Center for Molecular Biology, National Research and Innovation Agency, Cibinong, Bogor 16911, Indonesia; Master's Programme in Biomedical Sciences, Faculty of Medicine Universitas Indonesia, Jakarta 10430, Indonesia
| | - Andita Fitri Mutiara Rizki
- Eijkman Research Center for Molecular Biology, National Research and Innovation Agency, Cibinong, Bogor 16911, Indonesia; Master's Programme in Biomedical Sciences, Faculty of Medicine Universitas Indonesia, Jakarta 10430, Indonesia
| | - Yenny Djuardi
- Department of Parasitology, Faculty of Medicine Universitas Indonesia, Jakarta 10430, Indonesia
| | - I Made Artika
- Eijkman Research Center for Molecular Biology, National Research and Innovation Agency, Cibinong, Bogor 16911, Indonesia; Department of Biochemistry, Faculty of Mathematics and Natural Sciences, Bogor Agricultural University, Bogor 16680, Indonesia
| | - Josephine Elizabeth Siregar
- Eijkman Research Center for Molecular Biology, National Research and Innovation Agency, Cibinong, Bogor 16911, Indonesia.
| |
Collapse
|
12
|
Platon L, Baker DA, Ménard D. Modified Plasmodium falciparum Ring-Stage Survival Assay with ML10 Kinase Inhibitor. Antimicrob Agents Chemother 2023; 67:e0001723. [PMID: 37098950 PMCID: PMC10190288 DOI: 10.1128/aac.00017-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 04/10/2023] [Indexed: 04/27/2023] Open
Abstract
The ring-stage survival assay is the reference assay to measure in vitro Plasmodium falciparum artemisinin partial resistance. The main challenge of the standard protocol is to generate 0-to-3-h postinvasion ring stages (the stage least susceptible to artemisinin) from schizonts obtained by sorbitol treatment and Percoll gradient. We report here a modified protocol facilitating the production of synchronized schizonts when multiple strains are tested simultaneously, by using ML10, a protein kinase inhibitor, that reversibly blocks merozoite egress.
Collapse
Affiliation(s)
- Lucien Platon
- Institut Pasteur, Université Paris Cité, Malaria Genetics and Resistance Unit, INSERM U1201, Paris, France
- Sorbonne Université, Paris, France
- Institut Pasteur, Université Paris Cité, Malaria Parasite Biology and Vaccines Unit, Paris, France
| | - David A. Baker
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Didier Ménard
- Institut Pasteur, Université Paris Cité, Malaria Genetics and Resistance Unit, INSERM U1201, Paris, France
- Institut Pasteur, Université Paris Cité, Malaria Parasite Biology and Vaccines Unit, Paris, France
- Université de Strasbourg, Institute of Parasitology and Tropical Diseases, UR7292 Dynamics of Host-Pathogen Interactions, Strasbourg, France
- CHU Strasbourg, Laboratory of Parasitology and Medical Mycology, Strasbourg, France
| |
Collapse
|
13
|
Peter C, Annie R, Ward Kurt E, Jessica M, Rossarin S, Adeline CC, Kaneko O, Lin AH, Laurent R, Nadia A, Victor M, Julien L, Roger LG, Dennis K, Pablo B, Cook Gregory M, Georges S, Bruce R. Improving in vitro continuous cultivation of Plasmodium cynomolgi, a model for P. vivax. Parasitol Int 2022; 89:102589. [DOI: 10.1016/j.parint.2022.102589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/18/2022] [Accepted: 04/18/2022] [Indexed: 10/18/2022]
|
14
|
Assessment
in vitro
of the antimalarial and transmission blocking activities of Cipargamin and Ganaplacide in artemisinin resistant
Plasmodium falciparum. Antimicrob Agents Chemother 2022; 66:e0148121. [DOI: 10.1128/aac.01481-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Artemisinin resistance in
Plasmodium falciparum
has emerged and spread widely in the Greater Mekong Subregion threatening current first line artemisinin combination treatments. New antimalarial drugs are needed urgently. Cipargamin (KAE609) and ganaplacide (KAF156) are promising novel antimalarial compounds in advanced stages of development. Both compounds have potent asexual blood stage activities, inhibit
P. falciparum
gametocytogenesis and reduce oocyst development in anopheline mosquitoes. In this study, we compared the asexual and sexual stage activities of cipargamin, ganaplacide and artesunate in artemisinin resistant
P. falciparum
isolates (N=7, K13 mutation; C580Y, G449A and R539T) from Thailand and Cambodia. Asexual blood stage antimalarial activity was evaluated in a SYBR-green I based 72h
in vitro
assay, and the effects on male and female mature stage V gametocytes were assessed in the
P. falciparum
dual gamete formation assay. Ganaplacide had higher activities when compared to cipargamin and artesunate, with a mean (SD) IC50 against asexual stages of 5.5 (1.1) nM, 7.8 (3.9) nM for male gametocytes and 57.9 (59.6) nM for female gametocytes. Cipargamin had a similar potency against male and female gametocytes, with a mean (SD) IC50 of 123.1 (80.2) nM for male gametocytes, 88.5 (52.7) nM for female gametocytes and 2.4 (0.6) nM for asexual stages. Both cipargamin and ganaplacide showed significant transmission-blocking activities against artemisinin resistant
P. falciparum
in vitro
.
Collapse
|
15
|
Ong JJY, Russell B, Han JH. Plasmodium cynomolgi Berok Growth Inhibition Assay by Thiol-reactive Probe Based Flow Cytometric Measurement. Bio Protoc 2021; 11:e4147. [PMID: 34604452 DOI: 10.21769/bioprotoc.4147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 05/05/2021] [Accepted: 05/06/2021] [Indexed: 01/18/2023] Open
Abstract
The relapsing malaria species, Plasmodium vivax, is the most widely distributed and difficult-to-treat cause of human malaria. The merozoites of P. vivax preferentially invade ephemeral human CD71+ reticulocytes (nascent reticulocytes), thereby limiting the development of a robust continuous culture in vitro. Fortunately, P. vivax's sister species, P. cynomolgi Berok, can be cultured continuously, providing the ability to screen novel therapeutics drug and vaccine candidates in a reliable and high-throughput manner. Based on well-established growth inhibition activity (GIA) assays against P. falciparum and P. knowlesi, this protocol adopts the current flow cytometry assay methodology and investigates P. vivax inhibitory antibodies using the P. cynomolgi Berok invasion model based on the thiol-reactivity and DNA abundance of viable parasites in macaque erythrocytes. Established GIA assays screen antibodies at either a single concentration or high/low dose concentrations to provide quick insights for prioritizing potential antibodies capable of specifically interrupting parasite ligand and host receptor binding with minimal concentrations. Hence, this protocol expands on the existing GIA assay by using serially diluted antibodies and generating a dose-response curve to better quantify the inhibitory efficacy amongst selected vaccine candidates.
Collapse
Affiliation(s)
- Jessica Jie Ying Ong
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Bruce Russell
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Jin-Hee Han
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand.,Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
| |
Collapse
|
16
|
Restructured Mitochondrial-Nuclear Interaction in Plasmodium falciparum Dormancy and Persister Survival after Artemisinin Exposure. mBio 2021; 12:e0075321. [PMID: 34044591 PMCID: PMC8262848 DOI: 10.1128/mbio.00753-21] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Artemisinin and its semisynthetic derivatives (ART) are fast acting, potent antimalarials; however, their use in malaria treatment is frequently confounded by recrudescences from bloodstream Plasmodium parasites that enter into and later reactivate from a dormant persister state. Here, we provide evidence that the mitochondria of dihydroartemisinin (DHA)-exposed persisters are dramatically altered and enlarged relative to the mitochondria of young, actively replicating ring forms. Restructured mitochondrial-nuclear associations and an altered metabolic state are consistent with stress from reactive oxygen species. New contacts between the mitochondria and nuclei may support communication pathways of mitochondrial retrograde signaling, resulting in transcriptional changes in the nucleus as a survival response. Further characterization of the organelle communication and metabolic dependencies of persisters may suggest strategies to combat recrudescences of malaria after treatment.
Collapse
|
17
|
Montenegro LM, de Las Salas B, Neal AT, Tobon-Castaño A, Fairhurst RM, Lopera-Mesa TM. State of Artemisinin and Partner Drug Susceptibility in Plasmodium falciparum Clinical Isolates from Colombia. Am J Trop Med Hyg 2021; 104:263-270. [PMID: 33289466 DOI: 10.4269/ajtmh.20-0148] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Delayed parasite clearance time observed in Southeast Asia provided the first evidence of Plasmodium falciparum resistance to artemisinins. The ex vivo ring-stage survival assay (RSA) mimics parasite exposure to pharmacologically relevant artemisinin concentrations. Mutations in the C-terminal propeller domain of the putative kelch protein Pf3D7_1343700 (K13) are associated with artemisinin resistance. Variations in the pfmdr1 gene are associated with reduced susceptibility to the artemisinin partner drugs mefloquine (MQ) and lumefantrine (LF). To clarify the unknown landscape of artemisinin resistance in Colombia, 71 patients with uncomplicated P. falciparum malaria were enrolled in a non-randomized observational study in three endemic localities in 2014-2015. Each patient's parasite isolate was assessed for ex vivo RSA, K13-propeller mutations, pfmdr1 copy number, and pfmdr1 mutations at codons 86, 184, 1034, 1042, and 1246, associated with reduced susceptibility, and 50% inhibitory concentration (IC50) for other antimalarial drugs. Ex vivo RSAs were successful in 56% (40/71) of samples, and nine isolates showed survival rates > 1%. All isolates had wild-type K13-propeller sequences. All isolates harbored either of two pfmdr1 haplotypes, NFSDD (79.3%) and NFSDY (20.7%), and 7.1% of isolates had > 1 pfmdr1 gene. In vitro IC50 assays showed that variable proportions of isolates had decreased susceptibility to chloroquine (52.4%, > 100 nM), amodiaquine (31.2%, > 30 nM), MQ (34.3%, > 30 nM), and LF (3.2%, > 10 nM). In this study, we report ex vivo RSA and K13 data on P. falciparum isolates from Colombia. The identification of isolates with increased ex vivo RSA rates in the absence of K13-propeller mutations and no positivity at day three requires further investigation.
Collapse
Affiliation(s)
| | - Briegel de Las Salas
- 1Grupo Malaria, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Aaron T Neal
- 2Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland
| | | | - Rick M Fairhurst
- 2Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland
| | | |
Collapse
|
18
|
Mbye H, Bojang F, Jawara AS, Njie B, Mohammed NI, Okebe J, D'Alessandro U, Amambua-Ngwa A. Tolerance of Gambian Plasmodium falciparum to Dihydroartemisinin and Lumefantrine Detected by Ex Vivo Parasite Survival Rate Assay. Antimicrob Agents Chemother 2020; 65:e00720-20. [PMID: 33020162 PMCID: PMC7927851 DOI: 10.1128/aac.00720-20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 10/02/2020] [Indexed: 11/20/2022] Open
Abstract
Monitoring of Plasmodium falciparum sensitivity to antimalarial drugs in Africa is vital for malaria elimination. However, the commonly used ex vivo/in vitro 50% inhibitory concentration (IC50) test gives inconsistent results for several antimalarials, while the alternative ring-stage survival assay (RSA) for artemisinin derivatives has not been widely adopted. Here, we applied an alternative two-color flow cytometry-based parasite survival rate assay (PSRA) to detect ex vivo antimalarial tolerance in P. falciparum isolates from The Gambia. The PSRA infers parasite viability by quantifying reinvasion of uninfected cells following 3 consecutive days of drug exposure (10-fold the IC50 of drug for field isolates). The drug survival rate is obtained for each isolate from the slope of the growth/death curve. We obtained parasite survival rates of 41 isolates for dihydroartemisinin (DHA) and lumefantrine (LUM) out of 51 infections tested by ring-stage survival assay (RSA) against DHA. We also determined the genotypes for known drug resistance genetic loci in the P. falciparum genes Pfdhfr, Pfdhps, Pfmdr, Pfcrt, and Pfk13 The PSRA results determined for 41 Gambian isolates showed faster killing and lower variance after treatment with DHA than after treatment with LUM, despite a strong correlation between the two drugs. Four and three isolates were tolerant to DHA and LUM, respectively, with continuous growth during drug exposure. Isolates with the PfMDR1-Y184F mutant variant showed increased LUM survival, though the results were not statistically significant. Sulfadoxine/pyrimethamine (SP) resistance markers were fixed, while all other antimalarial variants were prevalent in more than 50% of the population. The PSRA detected ex vivo antimalarial tolerance in Gambian P. falciparum This calls for its wider application and for increased vigilance against resistance to artemisinin combination therapies (ACTs) in this population.
Collapse
Affiliation(s)
- Haddijatou Mbye
- West African Centre for Cell Biology of Infectious Pathogens, Department of Biochemistry, Cell and Molecular Biology, University of Ghana, Legon, Ghana
- MRC Unit The Gambia at London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Fatoumata Bojang
- MRC Unit The Gambia at London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Aminata Seedy Jawara
- MRC Unit The Gambia at London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | - Bekai Njie
- MRC Unit The Gambia at London School of Hygiene and Tropical Medicine, Fajara, The Gambia
| | | | - Joseph Okebe
- Department of International Public Health, Liverpool School of Tropical Medicine, United Kingdom
| | - Umberto D'Alessandro
- MRC Unit The Gambia at London School of Hygiene and Tropical Medicine, Fajara, The Gambia
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Alfred Amambua-Ngwa
- MRC Unit The Gambia at London School of Hygiene and Tropical Medicine, Fajara, The Gambia
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
19
|
Windle ST, Lane KD, Gadalla NB, Liu A, Mu J, Caleon RL, Rahman RS, Sá JM, Wellems TE. Evidence for linkage of pfmdr1, pfcrt, and pfk13 polymorphisms to lumefantrine and mefloquine susceptibilities in a Plasmodium falciparum cross. Int J Parasitol Drugs Drug Resist 2020; 14:208-217. [PMID: 33197753 PMCID: PMC7677662 DOI: 10.1016/j.ijpddr.2020.10.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 10/21/2020] [Accepted: 10/23/2020] [Indexed: 11/13/2022]
Abstract
BACKGROUND Lumefantrine and mefloquine are used worldwide in artemisinin-based combination therapy (ACT) of malaria. Better understanding of drug susceptibility and resistance is needed and can be obtained from studies of genetic crosses. METHODS Drug response phenotypes of a cross between Plasmodium falciparum lines 803 (Cambodia) and GB4 (Ghana) were obtained as half-maximal effective concentrations (EC50s) and days to recovery (DTR) after 24 h exposure to 500 nM lumefantrine. EC50s of mefloquine, halofantrine, chloroquine, and dihydroartemisinin were also determined. Quantitative trait loci (QTL) analysis and statistical tests with candidate genes were used to identify polymorphisms associated with response phenotypes. RESULTS Lumefantrine EC50s averaged 5.8-fold higher for the 803 than GB4 parent, and DTR results were 3-5 and 16-18 days, respectively. In 803 × GB4 progeny, outcomes of these two lumefantrine assays showed strong inverse correlation; these phenotypes also correlated strongly with mefloquine and halofantrine EC50s. By QTL analysis, lumefantrine and mefloquine phenotypes mapped to a chromosome 5 region containing codon polymorphisms N86Y and Y184F in the P. falciparum multidrug resistance 1 protein (PfMDR1). Statistical tests of candidate genes identified correlations between inheritance of PfK13 Kelch protein polymorphism C580Y (and possibly K189T) and lumefantrine and mefloquine susceptibilities. Correlations were detected between lumefantrine and chloroquine EC50s and polymorphisms N326S and I356T in the CVIET-type P. falciparum chloroquine resistance transporter (PfCRT) common to 803 and GB4. CONCLUSIONS Correlations in this study suggest common mechanisms of action in lumefantrine, mefloquine, and halofantrine responses. PfK13 as well as PfMDR1 and PfCRT polymorphisms may affect access and/or action of these arylaminoalcohol drugs at locations of hemoglobin digestion and heme metabolism. In endemic regions, pressure from use of lumefantrine or mefloquine in ACTs may drive selection of PfK13 polymorphisms along with versions of PfMDR1 and PfCRT associated with lower susceptibility to these drugs.
Collapse
Affiliation(s)
- Sean T Windle
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20892, USA
| | - Kristin D Lane
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20892, USA
| | - Nahla B Gadalla
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20892, USA
| | - Anna Liu
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20892, USA
| | - Jianbing Mu
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20892, USA
| | - Ramoncito L Caleon
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20892, USA
| | - Rifat S Rahman
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20892, USA
| | - Juliana M Sá
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20892, USA
| | - Thomas E Wellems
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20892, USA.
| |
Collapse
|
20
|
Clements RL, Streva V, Dumoulin P, Huang W, Owens E, Raj DK, Burleigh B, Llinás M, Winzeler EA, Zhang Q, Dvorin JD. A Novel Antiparasitic Compound Kills Ring-Stage Plasmodium falciparum and Retains Activity Against Artemisinin-Resistant Parasites. J Infect Dis 2020; 221:956-962. [PMID: 31616928 DOI: 10.1093/infdis/jiz534] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 10/11/2019] [Indexed: 11/14/2022] Open
Abstract
Spreading antimalarial resistance threatens effective treatment of malaria, an infectious disease caused by Plasmodium parasites. We identified a compound, BCH070, that inhibits asexual growth of multiple antimalarial-resistant strains of Plasmodium falciparum (half maximal inhibitory concentration [IC50] = 1-2 µM), suggesting that BCH070 acts via a novel mechanism of action. BCH070 preferentially kills early ring-form trophozoites, and, importantly, equally inhibits ring-stage survival of wild-type and artemisinin-resistant parasites harboring the PfKelch13:C580Y mutation. Metabolomic analysis demonstrates that BCH070 likely targets multiple pathways in the parasite. BCH070 is a promising lead compound for development of new antimalarial combination therapy that retains activity against artemisinin-resistant parasites.
Collapse
Affiliation(s)
- Rebecca L Clements
- Biological and Biomedical Sciences, Harvard Medical School, Boston, Massachusetts, USA.,Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Vincent Streva
- Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Peter Dumoulin
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Weigang Huang
- Division of Chemical Biology and Medicinal Chemistry, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Edward Owens
- Huck Center for Malaria Research, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Dipak K Raj
- Center for International Health Research, Rhode Island Hospital, Brown University Medical School, Providence, Rhode Island, USA
| | - Barbara Burleigh
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Manuel Llinás
- Huck Center for Malaria Research, Pennsylvania State University, University Park, Pennsylvania, USA.,Department of Chemistry, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Elizabeth A Winzeler
- School of Medicine, University of California San Diego, La Jolla, California, USA
| | - Qisheng Zhang
- Division of Chemical Biology and Medicinal Chemistry, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jeffrey D Dvorin
- Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts, USA.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
21
|
Portugaliza HP, Miyazaki S, Geurten FJ, Pell C, Rosanas-Urgell A, Janse CJ, Cortés A. Artemisinin exposure at the ring or trophozoite stage impacts Plasmodium falciparum sexual conversion differently. eLife 2020; 9:60058. [PMID: 33084568 PMCID: PMC7577739 DOI: 10.7554/elife.60058] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 10/07/2020] [Indexed: 12/12/2022] Open
Abstract
Malaria transmission is dependent on the formation of gametocytes in the human blood. The sexual conversion rate, the proportion of asexual parasites that convert into gametocytes at each multiplication cycle, is variable and reflects the relative parasite investment between transmission and maintaining the infection. The impact of environmental factors such as drugs on sexual conversion rates is not well understood. We developed a robust assay using gametocyte-reporter parasite lines to accurately measure the impact of drugs on sexual conversion rates, independently from their gametocytocidal activity. We found that exposure to subcurative doses of the frontline antimalarial drug dihydroartemisinin (DHA) at the trophozoite stage resulted in a ~ fourfold increase in sexual conversion. In contrast, no increase was observed when ring stages were exposed or in cultures in which sexual conversion was stimulated by choline depletion. Our results reveal a complex relationship between antimalarial drugs and sexual conversion, with potential public health implications.
Collapse
Affiliation(s)
- Harvie P Portugaliza
- ISGlobal, Hospital Clinic - Universitat de Barcelona, Barcelona, Spain.,Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium.,Department of Global Health, Amsterdam University Medical Centers, location Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Shinya Miyazaki
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Fiona Ja Geurten
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Christopher Pell
- Department of Global Health, Amsterdam University Medical Centers, location Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Amsterdam Institute for Global Health and Development (AIGHD), Amsterdam, Netherlands
| | - Anna Rosanas-Urgell
- Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Chris J Janse
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Alfred Cortés
- ISGlobal, Hospital Clinic - Universitat de Barcelona, Barcelona, Spain.,ICREA, Barcelona, Spain
| |
Collapse
|
22
|
A modified two-color flow cytometry assay to quantify in-vitro reinvasion and determine invasion phenotypes at low Plasmodium falciparum parasitemia. Exp Parasitol 2020; 218:107969. [PMID: 32858043 DOI: 10.1016/j.exppara.2020.107969] [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: 02/14/2020] [Revised: 07/31/2020] [Accepted: 08/05/2020] [Indexed: 11/22/2022]
Abstract
Invasion of human red blood cells (RBCs) by Plasmodium parasites is a crucial yet poorly characterised phenotype. Two-color flow cytometry (2cFCM) promises to be a very sensitive and high throughput method for phenotyping parasite invasion. However, current protocols require high (~1.0%) parasitemia for assay set-up and need to be adapted for low parasitemia samples, which are becoming increasingly common in low transmission settings. Background fluorescence from nuclei-containing uninfected RBCs and high autologous reinvasion rates (merozoite invasion of donor uninfected RBCs present at 50% assay volume) are some of the limitations to the method's sensitivity to enumerate low parasitemia (<0.5%) with nucleic acid-based stains. Here, we describe modifications for plating unlabeled donor to labeled target RBCs per assay well and for gating parasitemia, that produces accurate quantifications of low reinvasion parasitemia. Plasmodium falciparum 3D7, Dd2 and field isolates at various low and high parasitemia (0.05%-2.0%) were used to set-up SyBr Green 1-based 2cFCM invasion assays. Target RBCs were labeled with CTFR proliferation dye. We show that this dye combination allowed for efficient parasite invasion into target RBCs and that a 1:3 ratio of unlabeled to labeled RBCs per assay greatly skewed autologous reinvasion (p < 0.001). Accuracy of quantifying reinvasion was limited to an assay parasitemia of 0.02% with minimal background interference. Invasion inhibition by enzymatic treatments increased averagely by 10% (p<0.05) across the entire parasitemia range. The effect was greater for samples with <0.5% parasitemia. Overall, a more sensitive method for phenotyping invasion of low P. falciparum parasitemia is described.
Collapse
|
23
|
Davis SZ, Singh PP, Vendrely KM, Shoue DA, Checkley LA, McDew-White M, Button-Simons KA, Cassady Z, Sievert MAC, Foster GJ, Nosten FH, Anderson TJC, Ferdig MT. The extended recovery ring-stage survival assay provides a superior association with patient clearance half-life and increases throughput. Malar J 2020; 19:54. [PMID: 32005233 PMCID: PMC6995136 DOI: 10.1186/s12936-020-3139-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 01/24/2020] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Tracking and understanding artemisinin resistance is key for preventing global setbacks in malaria eradication efforts. The ring-stage survival assay (RSA) is the current gold standard for in vitro artemisinin resistance phenotyping. However, the RSA has several drawbacks: it is relatively low throughput, has high variance due to microscopy readout, and correlates poorly with the current benchmark for in vivo resistance, patient clearance half-life post-artemisinin treatment. Here a modified RSA is presented, the extended Recovery Ring-stage Survival Assay (eRRSA), using 15 cloned patient isolates from Southeast Asia with a range of patient clearance half-lives, including parasite isolates with and without kelch13 mutations. METHODS Plasmodium falciparum cultures were synchronized with single layer Percoll during the schizont stage of the intraerythrocytic development cycle. Cultures were left to reinvade to early ring-stage and parasitaemia was quantified using flow cytometry. Cultures were diluted to 2% haematocrit and 0.5% parasitaemia in a 96-well plate to start the assay, allowing for increased throughput and decreased variability between biological replicates. Parasites were treated with 700 nM of dihydroartemisinin or 0.02% dimethyl sulfoxide (DMSO) for 6 h, washed three times in drug-free media, and incubated for 66 or 114 h, when samples were collected and frozen for PCR amplification. A SYBR Green-based quantitative PCR method was used to quantify the fold-change between treated and untreated samples. RESULTS 15 cloned patient isolates from Southeast Asia with a range of patient clearance half-lives were assayed using the eRRSA. Due to the large number of pyknotic and dying parasites at 66 h post-exposure (72 h sample), parasites were grown for an additional cell cycle (114 h post-exposure, 120 h sample), which drastically improved correlation with patient clearance half-life compared to the 66 h post-exposure sample. A Spearman correlation of - 0.8393 between fold change and patient clearance half-life was identified in these 15 isolates from Southeast Asia, which is the strongest correlation reported to date. CONCLUSIONS eRRSA drastically increases the efficiency and accuracy of in vitro artemisinin resistance phenotyping compared to the traditional RSA, which paves the way for extensive in vitro phenotyping of hundreds of artemisinin resistant parasites.
Collapse
Affiliation(s)
- Sage Z Davis
- Eck Institute for Global Health, Dept. of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA.,Molecular, Cell, and Systems Biology Department, University of California Riverside, Riverside, CA, USA
| | - Puspendra P Singh
- Eck Institute for Global Health, Dept. of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
| | - Katelyn M Vendrely
- Eck Institute for Global Health, Dept. of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
| | - Douglas A Shoue
- Eck Institute for Global Health, Dept. of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
| | - Lisa A Checkley
- Eck Institute for Global Health, Dept. of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
| | | | - Katrina A Button-Simons
- Eck Institute for Global Health, Dept. of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
| | - Zione Cassady
- Eck Institute for Global Health, Dept. of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
| | - Mackenzie A C Sievert
- Eck Institute for Global Health, Dept. of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
| | - Gabriel J Foster
- Eck Institute for Global Health, Dept. of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
| | - François H Nosten
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Mae Sot, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine Research Building, University of Oxford Old Road Campus, Oxford, UK
| | | | - Michael T Ferdig
- Eck Institute for Global Health, Dept. of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA.
| |
Collapse
|
24
|
Artemisinin Bioactivity and Resistance in Malaria Parasites. Trends Parasitol 2019; 35:953-963. [DOI: 10.1016/j.pt.2019.09.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 09/13/2019] [Accepted: 09/13/2019] [Indexed: 11/22/2022]
|
25
|
Linzke M, Yan SLR, Tárnok A, Ulrich H, Groves MR, Wrenger C. Live and Let Dye: Visualizing the Cellular Compartments of the Malaria Parasite Plasmodium falciparum. Cytometry A 2019; 97:694-705. [PMID: 31738009 DOI: 10.1002/cyto.a.23927] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 10/03/2019] [Accepted: 10/24/2019] [Indexed: 12/15/2022]
Abstract
Malaria remains one of the deadliest diseases worldwide and it is caused by the protozoan parasite Plasmodium spp. Parasite visualization is an important tool for the correct detection of malarial cases but also to understand its biology. Advances in visualization techniques promote new insights into the complex life cycle and biology of Plasmodium parasites. Live cell imaging by fluorescence microscopy or flow cytometry are the foundation of the visualization technique for malaria research. In this review, we present an overview of possibilities in live cell imaging of the malaria parasite. We discuss some of the state-of-the-art techniques to visualize organelles and processes of the parasite and discuss limitation and advantages of each technique. © 2019 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Marleen Linzke
- Unit for Drug Discovery, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, Avenida Professor Lineu Prestes 1374, São Paulo, São Paulo, 05508-000, Brazil
| | - Sun Liu Rei Yan
- Unit for Drug Discovery, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, Avenida Professor Lineu Prestes 1374, São Paulo, São Paulo, 05508-000, Brazil
| | - Attila Tárnok
- Institute for Medical Informatics, Statistics and Epidemiology, Medical Faculty, University Leipzig, D-04107, Härtelstraße 16-18, Leipzig, Germany
| | - Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, Avenida Professor Lineu Prestes 748, São Paulo, São Paulo, 05508-900, Brazil
| | - Matthew R Groves
- Structural Biology Unit, Department of Pharmacy, Faculty of Science and Engineering, University of Groningen, 9713AV, Antonius Deusinglaan 1, AV Groningen, The Netherlands
| | - Carsten Wrenger
- Unit for Drug Discovery, Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, Avenida Professor Lineu Prestes 1374, São Paulo, São Paulo, 05508-000, Brazil
| |
Collapse
|
26
|
Plasmodium Genomics and Genetics: New Insights into Malaria Pathogenesis, Drug Resistance, Epidemiology, and Evolution. Clin Microbiol Rev 2019; 32:32/4/e00019-19. [PMID: 31366610 DOI: 10.1128/cmr.00019-19] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Protozoan Plasmodium parasites are the causative agents of malaria, a deadly disease that continues to afflict hundreds of millions of people every year. Infections with malaria parasites can be asymptomatic, with mild or severe symptoms, or fatal, depending on many factors such as parasite virulence and host immune status. Malaria can be treated with various drugs, with artemisinin-based combination therapies (ACTs) being the first-line choice. Recent advances in genetics and genomics of malaria parasites have contributed greatly to our understanding of parasite population dynamics, transmission, drug responses, and pathogenesis. However, knowledge gaps in parasite biology and host-parasite interactions still remain. Parasites resistant to multiple antimalarial drugs have emerged, while advanced clinical trials have shown partial efficacy for one available vaccine. Here we discuss genetic and genomic studies of Plasmodium biology, host-parasite interactions, population structures, mosquito infectivity, antigenic variation, and targets for treatment and immunization. Knowledge from these studies will advance our understanding of malaria pathogenesis, epidemiology, and evolution and will support work to discover and develop new medicines and vaccines.
Collapse
|
27
|
Dhingra SK, Gabryszewski SJ, Small-Saunders JL, Yeo T, Henrich PP, Mok S, Fidock DA. Global Spread of Mutant PfCRT and Its Pleiotropic Impact on Plasmodium falciparum Multidrug Resistance and Fitness. mBio 2019; 10:e02731-18. [PMID: 31040246 PMCID: PMC6495381 DOI: 10.1128/mbio.02731-18] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 03/25/2019] [Indexed: 12/12/2022] Open
Abstract
The global spread of Plasmodium falciparum chloroquine resistance transporter (PfCRT) variant haplotypes earlier caused the widespread loss of chloroquine (CQ) efficacy. In Asia, novel PfCRT mutations that emerged on the Dd2 allelic background have recently been implicated in high-level resistance to piperaquine, and N326S and I356T have been associated with genetic backgrounds in which resistance emerged to artemisinin derivatives. By analyzing large-scale genome sequencing data, we report that the predominant Asian CQ-resistant Dd2 haplotype is undetectable in Africa. Instead, the GB4 and previously unexplored Cam783 haplotypes predominate, along with wild-type, drug-sensitive PfCRT that has reemerged as the major haplotype. To interrogate how these alleles impact drug susceptibility, we generated pfcrt-modified isogenic parasite lines spanning the mutational interval between GB4 and Dd2, which includes Cam783 and involves amino acid substitutions at residues 326 and 356. Relative to Dd2, the GB4 and Cam783 alleles were observed to mediate lower degrees of resistance to CQ and the first-line drug amodiaquine, while resulting in higher growth rates. These findings suggest that differences in growth rates, a surrogate of parasite fitness, influence selection in the context of African infections that are frequently characterized by high transmission rates, mixed infections, increased immunity, and less recourse to treatment. We also observe that the Asian Dd2 allele affords partial protection against piperaquine yet does not directly impact artemisinin efficacy. Our results can help inform the regional recommendations of antimalarials, whose activity is influenced by and, in certain cases, enhanced against select PfCRT variant haplotypes.IMPORTANCE Our study defines the allelic distribution of pfcrt, an important mediator of multidrug resistance in Plasmodium falciparum, in Africa and Asia. We leveraged whole-genome sequence analysis and gene editing to demonstrate how current drug combinations can select different allelic variants of this gene and shape region-specific parasite population structures. We document the ability of PfCRT mutations to modulate parasite susceptibility to current antimalarials in dissimilar, pfcrt allele-specific ways. This study underscores the importance of actively monitoring pfcrt genotypes to identify emerging patterns of multidrug resistance and help guide region-specific treatment options.
Collapse
Affiliation(s)
- Satish K Dhingra
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, New York, USA
| | - Stanislaw J Gabryszewski
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, New York, USA
| | - Jennifer L Small-Saunders
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, New York, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Medical Center, New York, New York, USA
| | - Tomas Yeo
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, New York, USA
| | - Philipp P Henrich
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, New York, USA
| | - Sachel Mok
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, New York, USA
| | - David A Fidock
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, New York, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Medical Center, New York, New York, USA
| |
Collapse
|
28
|
Heller LE, Goggins E, Roepe PD. Dihydroartemisinin-Ferriprotoporphyrin IX Adduct Abundance in Plasmodium falciparum Malarial Parasites and the Relationship to Emerging Artemisinin Resistance. Biochemistry 2018; 57:6935-6945. [PMID: 30512926 DOI: 10.1021/acs.biochem.8b00960] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Previously (Heller, L. E., and Roepe, P. D. Quantification of Free Ferriprotoporphyrin IX Heme and Hemozoin for Artemisinin Sensitive versus Delayed Clearance Phenotype Plasmodium falciparum Malarial Parasites. Biochemistry, DOI: 10.1021/acs.biochem.8b00959, preceding paper in this issue), we quantified free ferriprotoporphyrin IX (FPIX) heme abundance for control versus delayed clearance phenotype (DCP) intraerythrocytic Plasmodium falciparum malarial parasites. Because artemisinin drugs are activated by free FPIX, these data predict that the abundance of long-hypothesized toxic artemisinin drug-FPIX covalent adducts might differ for control versus DCP parasites. If so, this would have important repercussions for understanding the mechanism of the DCP, also known as emerging artemisinin resistance. To test these predictions, we studied in vitro formation of FPIX-dihydroartemisinin (DHA) adducts and then for the first time quantified the abundance of FPIX-DHA adducts formed within live P. falciparum versus the stage of intraerythrocytic development. Using matched isogenic parasite strains, we quantified the adduct for DCP versus control parasite strains and found that mutant PfK13 mediates lower adduct abundance for DCP parasites. The results suggest improved models for the molecular pharmacology of artemisinin-based antimalarial drugs and the molecular mechanism of the DCP.
Collapse
Affiliation(s)
- Laura E Heller
- Department of Chemistry and Department of Biochemistry and Cellular and Molecular Biology , Georgetown University , 37th and O Streets Northwest , Washington, D.C. 20057 , United States
| | - Eibhlin Goggins
- Department of Chemistry and Department of Biochemistry and Cellular and Molecular Biology , Georgetown University , 37th and O Streets Northwest , Washington, D.C. 20057 , United States
| | - Paul D Roepe
- Department of Chemistry and Department of Biochemistry and Cellular and Molecular Biology , Georgetown University , 37th and O Streets Northwest , Washington, D.C. 20057 , United States
| |
Collapse
|
29
|
Breglio KF, Amato R, Eastman R, Lim P, Sa JM, Guha R, Ganesan S, Dorward DW, Klumpp-Thomas C, McKnight C, Fairhurst RM, Roberts D, Thomas C, Simon AK. A single nucleotide polymorphism in the Plasmodium falciparum atg18 gene associates with artemisinin resistance and confers enhanced parasite survival under nutrient deprivation. Malar J 2018; 17:391. [PMID: 30367653 PMCID: PMC6204056 DOI: 10.1186/s12936-018-2532-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 10/17/2018] [Indexed: 01/20/2023] Open
Abstract
Background Artemisinin-resistant Plasmodium falciparum has been reported throughout the Greater Mekong subregion and threatens to disrupt current malaria control efforts worldwide. Polymorphisms in kelch13 have been associated with clinical and in vitro resistance phenotypes; however, several studies suggest that the genetic determinants of resistance may involve multiple genes. Current proposed mechanisms of resistance conferred by polymorphisms in kelch13 hint at a connection to an autophagy-like pathway in P. falciparum. Results A SNP in autophagy-related gene 18 (atg18) was associated with long parasite clearance half-life in patients following artemisinin-based combination therapy. This gene encodes PfAtg18, which is shown to be similar to the mammalian/yeast homologue WIPI/Atg18 in terms of structure, binding abilities, and ability to form puncta in response to stress. To investigate the contribution of this polymorphism, the atg18 gene was edited using CRISPR/Cas9 to introduce a T38I mutation into a k13-edited Dd2 parasite. The presence of this SNP confers a fitness advantage by enabling parasites to grow faster in nutrient-limited settings. The mutant and parent parasites were screened against drug libraries of 6349 unique compounds. While the SNP did not modulate the parasite’s susceptibility to any of the anti-malarial compounds using a 72-h drug pulse, it did alter the parasite’s susceptibility to 227 other compounds. Conclusions These results suggest that the atg18 T38I polymorphism may provide additional resistance against artemisinin derivatives, but not partner drugs, even in the absence of kelch13 mutations, and may also be important in parasite survival during nutrient deprivation. Electronic supplementary material The online version of this article (10.1186/s12936-018-2532-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kimberly F Breglio
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA. .,Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| | - Roberto Amato
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Richard Eastman
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Pharath Lim
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Juliana M Sa
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Rajarshi Guha
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA.,Vertex Pharmaceuticals, Boston, MA, USA
| | - Sundar Ganesan
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - David W Dorward
- Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Carleen Klumpp-Thomas
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Crystal McKnight
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Rick M Fairhurst
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - David Roberts
- Radcliffe Department of Medicine, Medical Sciences Division, University of Oxford, Oxford, UK
| | - Craig Thomas
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Anna Katharina Simon
- Kennedy Institute of Rheumatology and Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
30
|
Kelch Mutations in Plasmodium falciparum Protein K13 Do Not Modulate Dormancy after Artemisinin Exposure and Sorbitol Selection In Vitro. Antimicrob Agents Chemother 2018; 62:AAC.02256-17. [PMID: 29463530 DOI: 10.1128/aac.02256-17] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 02/09/2018] [Indexed: 11/20/2022] Open
Abstract
Some Kelch mutations of the Plasmodium falciparum K13 protein confer increased survival to dihydroartemisinin (DHA)-treated ring-stage parasites. Here, we asked if K13 mutations affect a dormancy phenotype allowing parasites to survive DHA exposure and then sorbitol selection. Although recrudescence from dormancy differed between two distinct parasite lines, it was similar for isogenic lines carrying single-site substitutions in K13. Therefore, K13 mutations do not alter the DHA-sorbitol combined dormancy phenotype; rather, traits from other loci likely determine this phenotype.
Collapse
|
31
|
de Laurent ZR, Chebon LJ, Ingasia LA, Akala HM, Andagalu B, Ochola-Oyier LI, Kamau E. Polymorphisms in the K13 Gene in Plasmodium falciparum from Different Malaria Transmission Areas of Kenya. Am J Trop Med Hyg 2018; 98:1360-1366. [PMID: 29582728 DOI: 10.4269/ajtmh.17-0505] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
The development of artemisinin (ART)-resistant parasites in Southeast Asia (SEA) threatens malaria control globally. Mutations in the Kelch 13 (K13)-propeller domain have been useful in identifying ART resistance in SEA. ART combination therapy (ACT) remains highly efficacious in the treatment of uncomplicated malaria in Sub-Saharan Africa (SSA). However, it is crucial that the efficacy of ACT is closely monitored. Toward this effort, this study profiled the prevalence of K13 nonsynonymous mutations in different malaria ecological zones of Kenya and in different time periods, before (pre) and after (post) the introduction of ACT as the first-line treatment of malaria. Nineteen nonsynonymous mutations were present in the pre-ACT samples (N = 64) compared with 22 in the post-ACT samples (N = 251). Eight of these mutations were present in both pre- and post-ACT parasites. Interestingly, seven of the shared single-nucleotide polymorphisms were at higher frequencies in the pre-ACT than the post-ACT parasites. The A578S mutation reported in SSA and the V568G mutation reported in SEA were found in both pre- and post-ACT parasites, with their frequencies declining post-ACT. D584Y and R539K mutations were found only in post-ACT parasites; changes in these codons have also been reported in SEA with different amino acids. The N585K mutation described for the first time in this study was present only in post-ACT parasites, and it was the most prevalent mutation at a frequency of 5.2%. This study showed the type, prevalence, and frequency of K13 mutations that varied based on the malaria ecological zones and also between the pre- and post-ACT time periods.
Collapse
Affiliation(s)
- Zaydah R de Laurent
- Center for Biotechnology and Bioinformatics, University of Nairobi, Nairobi, Kenya.,Kenya Medical Research Institute/United States Army Medical Research Directorate-Kenya, Kisumu, Kenya
| | - Lorna J Chebon
- Kenya Medical Research Institute/United States Army Medical Research Directorate-Kenya, Kisumu, Kenya
| | - Luicer A Ingasia
- Kenya Medical Research Institute/United States Army Medical Research Directorate-Kenya, Kisumu, Kenya
| | - Hoseah M Akala
- Kenya Medical Research Institute/United States Army Medical Research Directorate-Kenya, Kisumu, Kenya
| | - Ben Andagalu
- Kenya Medical Research Institute/United States Army Medical Research Directorate-Kenya, Kisumu, Kenya
| | - Lynette Isabella Ochola-Oyier
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya.,Center for Biotechnology and Bioinformatics, University of Nairobi, Nairobi, Kenya
| | - Edwin Kamau
- Kenya Medical Research Institute/United States Army Medical Research Directorate-Kenya, Kisumu, Kenya.,Walter Reed National Military Medical Center (WRNMMC), Bethesda, Maryland
| |
Collapse
|
32
|
Zhang J, Feng GH, Zou CY, Su PC, Liu HE, Yang ZQ. Overview of the improvement of the ring-stage survival assay-a novel phenotypic assay for the detection of artemisinin-resistant Plasmodium falciparum. Zool Res 2018; 38:317-320. [PMID: 29280362 PMCID: PMC5767555 DOI: 10.24272/j.issn.2095-8137.2017.075] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Artemisinin resistance in Plasmodium falciparum threatens the remarkable efficacy of artemisinin-based combination therapies worldwide. Thus, greater insight into the resistance mechanism using monitoring tools is essential. The ring-stage survival assay is used for phenotyping artemisinin-resistance or decreased artemisinin sensitivity. Here, we review the progress of this measurement assay and explore its limitations and potential applications.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Pathogen Biology and Immunology, Kunming Medical University, Kunming Yunnan 650500, China
| | - Guo-Hua Feng
- Biomedical Engineering Research Center, Kunming Medical University, Kunming Yunnan 650500, China
| | - Chun-Yan Zou
- Guangxi Zhuang Autonomous Region People's Hospital, Nanning Guangxi 530021, China
| | - Pin-Can Su
- Transfusion Medicine Research Department, Yunnan Kunming Blood Center, Kunming Yunnan 650500, China
| | - Huai-E Liu
- Department of Pathogen Biology and Immunology, Kunming Medical University, Kunming Yunnan 650500, China.,Department of Infectious Diseases, The First Affiliated Hospital, Kunming Medical University, Kunming Yunnan 650032, China
| | - Zhao-Qing Yang
- Department of Pathogen Biology and Immunology, Kunming Medical University, Kunming Yunnan 650500, China.
| |
Collapse
|
33
|
Nsanzabana C, Djalle D, Guérin PJ, Ménard D, González IJ. Tools for surveillance of anti-malarial drug resistance: an assessment of the current landscape. Malar J 2018; 17:75. [PMID: 29422048 PMCID: PMC5806256 DOI: 10.1186/s12936-018-2185-9] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 01/10/2018] [Indexed: 12/12/2022] Open
Abstract
To limit the spread and impact of anti-malarial drug resistance and react accordingly, surveillance systems able to detect and track in real-time its emergence and spread need to be strengthened or in some places established. Currently, surveillance of anti-malarial drug resistance is done by any of three approaches: (1) in vivo studies to assess the efficacy of drugs in patients; (2) in vitro/ex vivo studies to evaluate parasite susceptibility to the drugs; and/or (3) molecular assays to detect validated gene mutations and/or gene copy number changes that are associated with drug resistance. These methods are complementary, as they evaluate different aspects of resistance; however, standardization of methods, especially for in vitro/ex vivo and molecular techniques, is lacking. The World Health Organization has developed a standard protocol for evaluating the efficacy of anti-malarial drugs, which is used by National Malaria Control Programmes to conduct their therapeutic efficacy studies. Regional networks, such as the East African Network for Monitoring Antimalarial Treatment and the Amazon Network for the Surveillance of Antimalarial Drug Resistance, have been set up to strengthen regional capacities for monitoring anti-malarial drug resistance. The Worldwide Antimalarial Resistance Network has been established to collate and provide global spatial and temporal trends information on the efficacy of anti-malarial drugs and resistance. While exchange of information across endemic countries is essential for monitoring anti-malarial resistance, sustainable funding for the surveillance and networking activities remains challenging. The technology landscape for molecular assays is progressing quite rapidly, and easy-to-use and affordable new techniques are becoming available. They also offer the advantage of high throughput analysis from a simple blood spots obtained from a finger prick. New technologies combined with the strengthening of national reference laboratories in malaria-endemic countries through standardized protocols and training plus the availability of a proficiency testing programme, would contribute to the improvement and sustainability of anti-malarial resistance surveillance networks worldwide.
Collapse
Affiliation(s)
| | - Djibrine Djalle
- Foundation for Innovative New Diagnostics (FIND), Geneva, Switzerland
| | - Philippe J Guérin
- WorldWide Antimalarial Resistance Network, Oxford, UK.,Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Didier Ménard
- Unité Biologie des Interactions Hôte-Parasite, Institut Pasteur, Paris, France
| | - Iveth J González
- Foundation for Innovative New Diagnostics (FIND), Geneva, Switzerland
| |
Collapse
|
34
|
Singh D, McPhee D, Paddon CJ, Cherry J, Maurya G, Mahale G, Patel Y, Kumar N, Singh S, Sharma B, Kushwaha L, Singh S, Kumar A. Amalgamation of Synthetic Biology and Chemistry for High-Throughput Nonconventional Synthesis of the Antimalarial Drug Artemisinin. Org Process Res Dev 2017. [DOI: 10.1021/acs.oprd.6b00414] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Dharmendra Singh
- Chemistry
Research and Development, Plot Number 123-AB, Ipca Laboratories Limited, Kandivali Industrial Estate, Kandivali
West, Mumbai 400067, India
| | - Derek McPhee
- Research
and Development, Amyris Inc., 5885 Hollis Street, Suite 100, Emeryville, California 94608, United States
| | - Christopher J. Paddon
- Research
and Development, Amyris Inc., 5885 Hollis Street, Suite 100, Emeryville, California 94608, United States
| | - Joel Cherry
- Research
and Development, Amyris Inc., 5885 Hollis Street, Suite 100, Emeryville, California 94608, United States
| | - Ghanshyam Maurya
- Chemistry
Research and Development, Plot Number 123-AB, Ipca Laboratories Limited, Kandivali Industrial Estate, Kandivali
West, Mumbai 400067, India
| | - Ganesh Mahale
- Chemistry
Research and Development, Plot Number 123-AB, Ipca Laboratories Limited, Kandivali Industrial Estate, Kandivali
West, Mumbai 400067, India
| | - Yogesh Patel
- Chemistry
Research and Development, Plot Number 123-AB, Ipca Laboratories Limited, Kandivali Industrial Estate, Kandivali
West, Mumbai 400067, India
| | - Neeraj Kumar
- Chemistry
Research and Development, Plot Number 123-AB, Ipca Laboratories Limited, Kandivali Industrial Estate, Kandivali
West, Mumbai 400067, India
| | - Subhash Singh
- Chemistry
Research and Development, Plot Number 123-AB, Ipca Laboratories Limited, Kandivali Industrial Estate, Kandivali
West, Mumbai 400067, India
| | - Brajesh Sharma
- Chemistry
Research and Development, Plot Number 123-AB, Ipca Laboratories Limited, Kandivali Industrial Estate, Kandivali
West, Mumbai 400067, India
| | - Lavkesh Kushwaha
- Chemistry
Research and Development, Plot Number 123-AB, Ipca Laboratories Limited, Kandivali Industrial Estate, Kandivali
West, Mumbai 400067, India
| | - Satinder Singh
- Chemistry
Research and Development, Plot Number 123-AB, Ipca Laboratories Limited, Kandivali Industrial Estate, Kandivali
West, Mumbai 400067, India
| | - Ashok Kumar
- Chemistry
Research and Development, Plot Number 123-AB, Ipca Laboratories Limited, Kandivali Industrial Estate, Kandivali
West, Mumbai 400067, India
| |
Collapse
|
35
|
Dama S, Niangaly H, Ouattara A, Sagara I, Sissoko S, Traore OB, Bamadio A, Dara N, Djimde M, Alhousseini ML, Goita S, Maiga H, Dara A, Doumbo OK, Djimde AA. Reduced ex vivo susceptibility of Plasmodium falciparum after oral artemether-lumefantrine treatment in Mali. Malar J 2017; 16:59. [PMID: 28148267 PMCID: PMC5289056 DOI: 10.1186/s12936-017-1700-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 01/18/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Artemisinin-based combination therapy is the recommended first-line treatment for uncomplicated falciparum malaria worldwide. However, recent studies conducted in Mali showed an increased frequency of recurrent parasitaemia following artemether-lumefantrine (AL) treatment. METHODS Study samples were collected during a large WANECAM study. Ex-vivo Plasmodium falciparum sensitivity to artemether and lumefantrine was assessed using the tritiated hypoxanthine-based assay. The prevalence of molecular markers of anti-malarial drug resistance (pfcrt K76T, pfmdr1 N86Y and K13-propeller) were measured by PCR and/or sequencing. RESULTS Overall 61 samples were successfully analysed in ex vivo studies. Mean IC50s increased significantly between baseline and recurrent parasites for both artemether (1.6 nM vs 3.2 nM, p < 0.001) and lumefantrine (1.4 nM vs 3.4 nM, p = 0.004). Wild type Pfmdr1 N86 allele was selected after treatment (71 vs 91%, 112 of 158 vs 95 of 105, p < 0.001) but not the wild type pfcrt K76 variant (23.5 vs 24.8%, 40 of 170 vs 26 of 105, p = 0.9). Three non-synonymous K13-propeller SNPs (A522C, A578S, and G638R) were found with allele frequencies <2%. CONCLUSION Malian post-AL P. falciparum isolates were less susceptible to artemether and lumefantrine than baseline isolates.
Collapse
Affiliation(s)
- Souleymane Dama
- Malaria Research and Training Center, Faculty of Pharmacy and Faculty of Medicine and Dentistry, University of Sciences, Technique and Technology of Bamako, P.O. Box 1805, Bamako, Mali
| | - Hamidou Niangaly
- Malaria Research and Training Center, Faculty of Pharmacy and Faculty of Medicine and Dentistry, University of Sciences, Technique and Technology of Bamako, P.O. Box 1805, Bamako, Mali
| | - Amed Ouattara
- Malaria Research and Training Center, Faculty of Pharmacy and Faculty of Medicine and Dentistry, University of Sciences, Technique and Technology of Bamako, P.O. Box 1805, Bamako, Mali
| | - Issaka Sagara
- Malaria Research and Training Center, Faculty of Pharmacy and Faculty of Medicine and Dentistry, University of Sciences, Technique and Technology of Bamako, P.O. Box 1805, Bamako, Mali
| | - Sekou Sissoko
- Malaria Research and Training Center, Faculty of Pharmacy and Faculty of Medicine and Dentistry, University of Sciences, Technique and Technology of Bamako, P.O. Box 1805, Bamako, Mali
| | - Oumar Bila Traore
- Malaria Research and Training Center, Faculty of Pharmacy and Faculty of Medicine and Dentistry, University of Sciences, Technique and Technology of Bamako, P.O. Box 1805, Bamako, Mali
| | - Amadou Bamadio
- Malaria Research and Training Center, Faculty of Pharmacy and Faculty of Medicine and Dentistry, University of Sciences, Technique and Technology of Bamako, P.O. Box 1805, Bamako, Mali
| | - Niawanlou Dara
- Malaria Research and Training Center, Faculty of Pharmacy and Faculty of Medicine and Dentistry, University of Sciences, Technique and Technology of Bamako, P.O. Box 1805, Bamako, Mali
| | - Moussa Djimde
- Malaria Research and Training Center, Faculty of Pharmacy and Faculty of Medicine and Dentistry, University of Sciences, Technique and Technology of Bamako, P.O. Box 1805, Bamako, Mali
| | - Mohamed Lamine Alhousseini
- Malaria Research and Training Center, Faculty of Pharmacy and Faculty of Medicine and Dentistry, University of Sciences, Technique and Technology of Bamako, P.O. Box 1805, Bamako, Mali
| | - Siaka Goita
- Malaria Research and Training Center, Faculty of Pharmacy and Faculty of Medicine and Dentistry, University of Sciences, Technique and Technology of Bamako, P.O. Box 1805, Bamako, Mali
| | - Hamma Maiga
- Malaria Research and Training Center, Faculty of Pharmacy and Faculty of Medicine and Dentistry, University of Sciences, Technique and Technology of Bamako, P.O. Box 1805, Bamako, Mali
| | - Antoine Dara
- Malaria Research and Training Center, Faculty of Pharmacy and Faculty of Medicine and Dentistry, University of Sciences, Technique and Technology of Bamako, P.O. Box 1805, Bamako, Mali
| | - Ogobara K. Doumbo
- Malaria Research and Training Center, Faculty of Pharmacy and Faculty of Medicine and Dentistry, University of Sciences, Technique and Technology of Bamako, P.O. Box 1805, Bamako, Mali
| | - Abdoulaye A. Djimde
- Malaria Research and Training Center, Faculty of Pharmacy and Faculty of Medicine and Dentistry, University of Sciences, Technique and Technology of Bamako, P.O. Box 1805, Bamako, Mali
| |
Collapse
|
36
|
Ex Vivo Maturation Assay for Testing Antimalarial Sensitivity of Rodent Malaria Parasites. Antimicrob Agents Chemother 2016; 60:6859-6866. [PMID: 27600050 DOI: 10.1128/aac.01292-16] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2016] [Accepted: 08/30/2016] [Indexed: 11/20/2022] Open
Abstract
Ex vivo assay systems provide a powerful approach to studying human malaria parasite biology and to testing antimalarials. For rodent malaria parasites, short-term in vitro culture and ex vivo antimalarial susceptibility assays are relatively cumbersome, relying on in vivo passage for synchronization, since ring-stage parasites are an essential starting material. Here, we describe a new approach based on the enrichment of ring-stage Plasmodium berghei, P. yoelii, and P. vinckei vinckei using a single-step Percoll gradient. Importantly, we demonstrate that the enriched ring-stage parasites develop synchronously regardless of the parasite strain or species used. Using a flow cytometry assay with Hoechst and ethidium or MitoTracker dye, we show that parasite development is easily and rapidly monitored. Finally, we demonstrate that this approach can be used to screen antimalarial drugs.
Collapse
|
37
|
Endoperoxide Drug Cross-Resistance Patterns for Plasmodium falciparum Exhibiting an Artemisinin Delayed-Clearance Phenotype. Antimicrob Agents Chemother 2016; 60:6952-6956. [PMID: 27600038 DOI: 10.1128/aac.00857-16] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 08/28/2016] [Indexed: 11/20/2022] Open
Abstract
The ring-stage susceptibility assay was modified to quantify the susceptibilities of multiple strains of control and delayed-clearance phenotype (DCP) Plasmodium falciparum strains to seven endoperoxide antimalarial drugs. The susceptibility of all of the DCP lines to six of the drugs was lower than that of the controls. In contrast, DCP parasites did not show reduced susceptibility to the synthetic endoperoxide drug OZ439. These data show that it is possible to circumvent emerging artemisinin resistance with a modified endoperoxide drug.
Collapse
|
38
|
Woodrow CJ, White NJ. The clinical impact of artemisinin resistance in Southeast Asia and the potential for future spread. FEMS Microbiol Rev 2016; 41:34-48. [PMID: 27613271 PMCID: PMC5424521 DOI: 10.1093/femsre/fuw037] [Citation(s) in RCA: 151] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 04/11/2016] [Accepted: 07/31/2016] [Indexed: 11/25/2022] Open
Abstract
Artemisinins are the most rapidly acting of currently available antimalarial drugs. Artesunate has become the treatment of choice for severe malaria, and artemisinin-based combination therapies (ACTs) are the foundation of modern falciparum malaria treatment globally. Their safety and tolerability profile is excellent. Unfortunately, Plasmodium falciparum infections with mutations in the ‘K13’ gene, with reduced ring-stage susceptibility to artemisinins, and slow parasite clearance in patients treated with ACTs, are now widespread in Southeast Asia. We review clinical efficacy data from the region (2000–2015) that provides strong evidence that the loss of first-line ACTs in western Cambodia, first artesunate-mefloquine and then DHA-piperaquine, can be attributed primarily to K13 mutated parasites. The ring-stage activity of artemisinins is therefore critical for the sustained efficacy of ACTs; once it is lost, rapid selection of partner drug resistance and ACT failure are inevitable consequences. Consensus methods for monitoring artemisinin resistance are now available. Despite increased investment in regional control activities, ACTs are failing across an expanding area of the Greater Mekong subregion. Although multiple K13 mutations have arisen independently, successful multidrug-resistant parasite genotypes are taking over and threaten to spread to India and Africa. Stronger containment efforts and new approaches to sustaining long-term efficacy of antimalarial regimens are needed to prevent a global malaria emergency. Artemisinin resistance in Plasmodium falciparum malaria is causing failure of artemisinin-based combination therapies across an expanding area of Southeast Asia, undermining control and elimination efforts. The potential global consequences can only be avoided by new approaches that ensure sustained efficacy for antimalarial regimens in malaria affected populations.
Collapse
Affiliation(s)
- Charles J Woodrow
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, 420/6, Rajvithi Road, Bangkok 10400, Thailand
| | - Nicholas J White
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, 420/6, Rajvithi Road, Bangkok 10400, Thailand
| |
Collapse
|
39
|
Linares M, Viera S, Crespo B, Franco V, Gómez-Lorenzo MG, Jiménez-Díaz MB, Angulo-Barturen Í, Sanz LM, Gamo FJ. Identifying rapidly parasiticidal anti-malarial drugs using a simple and reliable in vitro parasite viability fast assay. Malar J 2015; 14:441. [PMID: 26542470 PMCID: PMC4635989 DOI: 10.1186/s12936-015-0962-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 10/22/2015] [Indexed: 11/21/2022] Open
Abstract
Background The emergence of Plasmodium falciparum resistance to artemisinins threatens to undermine the effectiveness of artemisinin-based combination anti-malarial therapy. Developing suitable drugs to replace artemisinins requires the identification of new compounds that display rapid parasite killing kinetics. However, no current methods fully meet the requirements to screen
large compound libraries for candidates with such properties. This study describes the development and validation of an in vitro parasite viability fast assay for identifying rapidly parasiticidal anti-malarial drugs. Methods Parasite killing kinetics were determined by first culturing unlabelled erythrocytes with P. falciparum in the presence of anti-malarial drugs for 24 or 48 h. After removing the drug, samples were added to erythrocytes pre-labelled with intracellular dye to allow their subsequent identification. The ability of viable parasites to re-establish infection in labelled erythrocytes could then be detected by two-colour flow cytometry after tagging of parasite DNA. Thus, double-stained erythrocytes (with the pre-labelled intracellular dye and the parasite DNA dye) result only after establishment of new infections by surviving parasites. The capacity of the test anti-malarial drugs to eliminate viable parasites within 24 or 48 h could, therefore, be determined. Results The parasite viability fast assay could be completed within 48 h following drug treatment and distinguished between rapidly parasiticidal anti-malarial drugs versus those acting more slowly. The assay was validated against ten standard anti-malarial agents with known properties and results correlated well with established methods. An abbreviated assay, suitable for adaption to medium–high throughput screening, was validated and applied against a set of 20 compounds retrieved from the publically available Medicines for Malaria Venture ‘Malaria Box’. Conclusion The quantification of new infections to determine parasite viability offers important advantages over existing methods, and is amenable to medium–high throughput screening. In particular, the parasite viability fast assay allows discrimination of rapidly parasiticidal anti-malarial candidates.
Collapse
Affiliation(s)
- María Linares
- R&D Alternative Discovery and Development, Diseases of the Developing World, GlaxoSmithKline, 28760 Tres Cantos, Madrid, Spain.
| | - Sara Viera
- R&D Alternative Discovery and Development, Diseases of the Developing World, GlaxoSmithKline, 28760 Tres Cantos, Madrid, Spain.
| | - Benigno Crespo
- R&D Alternative Discovery and Development, Diseases of the Developing World, GlaxoSmithKline, 28760 Tres Cantos, Madrid, Spain.
| | - Virginia Franco
- R&D Alternative Discovery and Development, Diseases of the Developing World, GlaxoSmithKline, 28760 Tres Cantos, Madrid, Spain.
| | - María G Gómez-Lorenzo
- R&D Alternative Discovery and Development, Diseases of the Developing World, GlaxoSmithKline, 28760 Tres Cantos, Madrid, Spain.
| | - María Belén Jiménez-Díaz
- R&D Alternative Discovery and Development, Diseases of the Developing World, GlaxoSmithKline, 28760 Tres Cantos, Madrid, Spain.
| | - Íñigo Angulo-Barturen
- R&D Alternative Discovery and Development, Diseases of the Developing World, GlaxoSmithKline, 28760 Tres Cantos, Madrid, Spain.
| | - Laura María Sanz
- R&D Alternative Discovery and Development, Diseases of the Developing World, GlaxoSmithKline, 28760 Tres Cantos, Madrid, Spain.
| | - Francisco-Javier Gamo
- R&D Alternative Discovery and Development, Diseases of the Developing World, GlaxoSmithKline, 28760 Tres Cantos, Madrid, Spain.
| |
Collapse
|
40
|
St Laurent B, Miller B, Burton TA, Amaratunga C, Men S, Sovannaroth S, Fay MP, Miotto O, Gwadz RW, Anderson JM, Fairhurst RM. Artemisinin-resistant Plasmodium falciparum clinical isolates can infect diverse mosquito vectors of Southeast Asia and Africa. Nat Commun 2015; 6:8614. [PMID: 26485448 PMCID: PMC4616032 DOI: 10.1038/ncomms9614] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 09/10/2015] [Indexed: 11/17/2022] Open
Abstract
Artemisinin-resistant Plasmodium falciparum parasites are rapidly spreading in Southeast Asia, yet nothing is known about their transmission. This knowledge gap and the possibility that these parasites will spread to Africa endanger global efforts to eliminate malaria. Here we produce gametocytes from parasite clinical isolates that displayed artemisinin resistance in patients and in vitro, and use them to infect native and non-native mosquito vectors. We show that contemporary artemisinin-resistant isolates from Cambodia develop and produce sporozoites in two Southeast Asian vectors, Anopheles dirus and Anopheles minimus, and the major African vector, Anopheles coluzzii (formerly Anopheles gambiae M). The ability of artemisinin-resistant parasites to infect such highly diverse Anopheles species, combined with their higher gametocyte prevalence in patients, may explain the rapid expansion of these parasites in Cambodia and neighbouring countries, and further compromise efforts to prevent their global spread. It is unknown whether artemisinin-resistant malaria parasites from Southeast Asia can infect any African species of Anopheles mosquitoes and thus spread to Africa. Here, St. Laurent et al. show that artemisinin-resistant isolates from Cambodia can indeed infect the major African vector, Anopheles coluzzii.
Collapse
Affiliation(s)
- Brandyce St Laurent
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway, Rockville, Maryland 20852, USA
| | - Becky Miller
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway, Rockville, Maryland 20852, USA
| | - Timothy A Burton
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway, Rockville, Maryland 20852, USA
| | - Chanaki Amaratunga
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway, Rockville, Maryland 20852, USA
| | - Sary Men
- National Center for Parasitology, Entomology and Malaria Control, Phnom Penh 12101, Cambodia
| | - Siv Sovannaroth
- National Center for Parasitology, Entomology and Malaria Control, Phnom Penh 12101, Cambodia
| | - Michael P Fay
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland 20852, USA
| | - Olivo Miotto
- Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand.,Malaria Programme, Wellcome Trust Sanger Institute, Hinxton CB10 1SA, UK.,Medical Research Council (MRC) Centre for Genomics and Global Health, University of Oxford, Oxford OX3 7BN, UK
| | - Robert W Gwadz
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway, Rockville, Maryland 20852, USA
| | - Jennifer M Anderson
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway, Rockville, Maryland 20852, USA
| | - Rick M Fairhurst
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 12735 Twinbrook Parkway, Rockville, Maryland 20852, USA
| |
Collapse
|
41
|
Rebelo M, Tempera C, Fernandes JF, Grobusch MP, Hänscheid T. Assessing anti-malarial drug effects ex vivo using the haemozoin detection assay. Malar J 2015; 14:140. [PMID: 25879757 PMCID: PMC4393616 DOI: 10.1186/s12936-015-0657-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Accepted: 03/17/2015] [Indexed: 11/20/2022] Open
Abstract
Background In vitro sensitivity assays are crucial to detect and monitor drug resistance. Plasmodium falciparum has developed resistance to almost all anti-malarial drugs. Although different in vitro drug assays are available, some of their inherent characteristics limit their application, especially in the field. A recently developed approach based on the flow cytometric detection of haemozoin (Hz) allowed reagent-free monitoring of parasite maturation and detection of drug effects in culture-adapted parasites. In this study, the set-up, performance and usefulness of this novel assay were investigated under field conditions in Gabon. Methods An existing flow cytometer (Cyflow Blue) was modified on site to detect light depolarization caused by Hz. Blood from malaria patients was incubated for 72 hrs with increasing concentrations of chloroquine, artesunate and artemisinin. The percentage of depolarizing red blood cells (RBC) was used as maturation indicator and measured at 24, 48 and 72 hrs of incubation to determine parasite growth and drug effects. Results The flow cytometer was easily adapted on site to detect light depolarization caused by Hz. Analysis of ex vivo cultures of parasites, obtained from blood samples of malaria patients, showed four different growth profiles. In 39/46 samples, 50% inhibitory concentrations (IC50) were successfully determined. IC50 values for chloroquine were higher than 200 nM in 70% of the samples, indicating the presence of chloroquine-resistant parasites. For artesunate and artemisinin, IC50 values ranged from 0.9 to 60 nM and from 2.2 nM to 124 nM, respectively, indicating fully sensitive parasites. Conclusion Flow cytometric detection of Hz allowed the detection of drug effects in blood samples from malaria patients, without using additional reagents or complex protocols. Adjustment of the initial parasitaemia was not required, which greatly simplifies the protocol, although it may lead to different IC50 values. Further investigation of set-up conditions of the Hz assay, as well as future studies in various settings should be performed to further determine the usefulness of this assay as a tool for rapid resistance testing in malaria-endemic countries.
Collapse
Affiliation(s)
- Maria Rebelo
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Av Prof Egas Moniz, Lisbon, P-1649-028, Portugal. .,Centre de Recherches Médicales de Lambaréné - CERMEL, Albert Schweitzer Hospital, Lambaréné, Gabon.
| | - Carolina Tempera
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Av Prof Egas Moniz, Lisbon, P-1649-028, Portugal.
| | - José F Fernandes
- Centre de Recherches Médicales de Lambaréné - CERMEL, Albert Schweitzer Hospital, Lambaréné, Gabon. .,Institut für Tropenmedizin, Universität Tübingen, Tübingen, Germany. .,Centre of Tropical Medicine and Travel Medicine, Amsterdam Medical Centre, University of Amsterdam, Amsterdam, The Netherlands.
| | - Martin P Grobusch
- Centre de Recherches Médicales de Lambaréné - CERMEL, Albert Schweitzer Hospital, Lambaréné, Gabon. .,Institut für Tropenmedizin, Universität Tübingen, Tübingen, Germany. .,Centre of Tropical Medicine and Travel Medicine, Amsterdam Medical Centre, University of Amsterdam, Amsterdam, The Netherlands.
| | - Thomas Hänscheid
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Av Prof Egas Moniz, Lisbon, P-1649-028, Portugal. .,Centre de Recherches Médicales de Lambaréné - CERMEL, Albert Schweitzer Hospital, Lambaréné, Gabon. .,Instituto de Microbiologia, Faculdade de Medicina, Lisbon, Portugal.
| |
Collapse
|
42
|
Rebelo M, Tempera C, Bispo C, Andrade C, Gardner R, Shapiro HM, Hänscheid T. Light depolarization measurements in malaria: A new job for an old friend. Cytometry A 2015; 87:437-45. [PMID: 25808846 DOI: 10.1002/cyto.a.22659] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Revised: 01/28/2015] [Accepted: 02/27/2015] [Indexed: 12/15/2022]
Abstract
The use of flow cytometry in malaria research has increased over the last decade. Most approaches use nucleic acid stains to detect parasite DNA and RNA and require complex multi-color, multi-parameter analysis to reliably detect infected red blood cells (iRBCs). We recently described a novel and simpler approach to parasite detection based on flow cytometric measurement of scattered light depolarization caused by hemozoin (Hz), a pigment formed by parasite digestion of hemoglobin in iRBCs. Depolarization measurement by flow cytometry was described in 1987; however, patent issues restricted its use to a single manufacturer's hematology analyzers until 2009. Although we recently demonstrated that depolarization measurement of Hz, easily implemented on a bench top flow cytometer (Cyflow), provided useful information for malaria work, doubts regarding its application and utility remain in both the flow cytometry and malaria communities, at least in part because instrument manufacturers do not offer the option of measuring depolarized scatter. Under such circumstances, providing other researchers with guidance as to how to do this seemed to offer the most expeditious way to resolve the issue. We accordingly examined how several commercially available flow cytometers (CyFlow SL, MoFLo, Attune and Accuri C6) could be modified to detect depolarization due to the presence of free Hz on solution, or of Hz in leukocytes or erythrocytes from rodent or human blood. All were readily adapted, with substantially equivalent results obtained with lasers emitting over a wide wavelength range. Other instruments now available may also be modifiable for Hz measurement. Cytometric detection of Hz using depolarization is useful to study different aspects of malaria. Adding additional parameters, such as DNA content and base composition and RNA content, can demonstrably provide improved accuracy and sensitivity of parasite detection and characterization, allowing malaria researchers and eventually clinicians to benefit from cytometric technology.
Collapse
Affiliation(s)
- Maria Rebelo
- Molecular Microbiology and Infection Unit, Instituto De Medicina Molecular, Faculdade De Medicina, Lisbon, Portugal
| | | | | | | | | | | | | |
Collapse
|
43
|
Antimalarial action of artesunate involves DNA damage mediated by reactive oxygen species. Antimicrob Agents Chemother 2014; 59:317-25. [PMID: 25348537 DOI: 10.1128/aac.03663-14] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Artemisinin-based combination therapy (ACT) is the recommended first-line treatment for Plasmodium falciparum malaria. It has been suggested that the cytotoxic effect of artemisinin is mediated by free radicals followed by the alkylation of P. falciparum proteins. The endoperoxide bridge, the active moiety of artemisinin derivatives, is cleaved in the presence of ferrous iron, generating reactive oxygen species (ROS) and other free radicals. However, the emergence of resistance to artemisinin in P. falciparum underscores the need for new insights into the molecular mechanisms of antimalarial activity of artemisinin. Here we show that artesunate (ART) induces DNA double-strand breaks in P. falciparum in a physiologically relevant dose- and time-dependent manner. DNA damage induced by ART was accompanied by an increase in the intracellular ROS level in the parasites. Mannitol, a ROS scavenger, reversed the cytotoxic effect of ART and reduced DNA damage, and modulation of glutathione (GSH) levels was found to impact ROS and DNA damage induced by ART. Accumulation of ROS, increased DNA damage, and the resulting antiparasite effect suggest a causal relationship between ROS, DNA damage, and parasite death. Finally, we also show that ART-induced ROS production involves a potential role for NADPH oxidase, an enzyme involved in the production of superoxide anions. Our results with P. falciparum provide novel insights into previously unknown molecular mechanisms underlying the antimalarial activity of artemisinin derivatives and may help in the design of next-generation antimalarial drugs against the most virulent Plasmodium species.
Collapse
|