1
|
Sakura T, Ishii R, Yoshida E, Kita K, Kato T, Inaoka DK. Accelerating Antimalarial Drug Discovery with a New High-Throughput Screen for Fast-Killing Compounds. ACS Infect Dis 2024; 10:4115-4126. [PMID: 39561299 DOI: 10.1021/acsinfecdis.4c00328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
The urgent need for rapidly acting compounds in the development of antimalarial drugs underscores the significance of such compounds in overcoming resistance issues and improving patient adherence to antimalarial treatments. The present study introduces a high-throughput screening (HTS) approach using 1536-well plates, employing Plasmodium falciparum lactate dehydrogenase (PfLDH) combined with nitroreductase (NTR) and fluorescent probes to evaluate inhibition of the growth of the asexual blood stage of malaria parasites. This method was adapted to efficiently assess the speed of action profiling (SAP) in a 384-well plate format, streamlining the traditionally time-consuming screening process. By successfully screening numerous compounds, this approach identified fast-killing hits early in the screening process, addressing challenges associated with artemisinin-based combination therapies. The high-throughput SAP method is expected to be of value in continuously monitoring fast-killing properties during structure-activity relationship studies, expediting the identification and development of novel, rapidly acting antimalarial drugs within phenotypic drug discovery campaigns.
Collapse
Affiliation(s)
- Takaya Sakura
- Department of Molecular Infection Dynamics, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki 852-8523, Japan
- School of Tropical Medicine and Global Health, Nagasaki University, Sakamoto, Nagasaki 852-8523, Japan
| | - Ryuta Ishii
- Department of Cellular Architecture Studies, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki 852-8523, Japan
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co., Ltd., Osaka 561-0825, Japan
| | - Eri Yoshida
- Department of Molecular Infection Dynamics, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki 852-8523, Japan
| | - Kiyoshi Kita
- School of Tropical Medicine and Global Health, Nagasaki University, Sakamoto, Nagasaki 852-8523, Japan
- Department of Infection Biochemistry, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki 852-8523, Japan
- Department of Biomedical Chemistry, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Teruhisa Kato
- Laboratory for Drug Discovery and Disease Research, Shionogi & Co., Ltd., Osaka 561-0825, Japan
- Exploratory Research for Drug Discovery, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki 852-8523, Japan
| | - Daniel Ken Inaoka
- Department of Molecular Infection Dynamics, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki 852-8523, Japan
- School of Tropical Medicine and Global Health, Nagasaki University, Sakamoto, Nagasaki 852-8523, Japan
- Department of Infection Biochemistry, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki 852-8523, Japan
- Department of Biomedical Chemistry, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| |
Collapse
|
2
|
Ullah I, Farringer MA, Burkhard AY, Hathaway E, Khushu M, Willett BC, Shin SH, Sharma AI, Martin MC, Shao KL, Dvorin JD, Hartl DL, Volkman SK, Bopp S, Absalon S, Wirth DF. Artemisinin resistance mutations in Pfcoronin impede hemoglobin uptake. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.22.572193. [PMID: 38187525 PMCID: PMC10769401 DOI: 10.1101/2023.12.22.572193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Artemisinin (ART) combination therapies have been critical in reducing malaria morbidity and mortality, but these important drugs are threatened by growing resistance associated with mutations in Pfcoronin and Pfkelch13 . Here, we describe the mechanism of Pfcoronin -mediated ART resistance. Pf Coronin interacts with Pf Actin and localizes to the parasite plasma membrane (PPM), the digestive vacuole (DV) membrane, and membrane of a newly identified preDV compartment-all structures involved in the trafficking of hemoglobin from the RBC for degradation in the DV. Pfcoronin mutations alter Pf Actin homeostasis and impair the development and morphology of the preDV. Ultimately, these changes are associated with decreased uptake of red blood cell cytosolic contents by ring-stage Plasmodium falciparum . Previous work has identified decreased hemoglobin uptake as the mechanism of Pfkelch 13-mediated ART resistance. This work demonstrates that Pf Coronin appears to act via a parallel pathway. For both Pfkelch13 -mediated and Pfcoronin -mediated ART resistance, we hypothesize that the decreased hemoglobin uptake in ring stage parasites results in less heme-based activation of the artemisinin endoperoxide ring and reduced cytocidal activity. This study deepens our understanding of ART resistance, as well as hemoglobin uptake and development of the DV in early-stage parasites.
Collapse
|
3
|
Lima C, Verdaguer IB, Wunderlich G, Katzin AM, Crabb BS, Gilson PR, Azevedo MF. Conditional expression of NanoLuc luciferase through a multimodular system offers rapid detection of antimalarial drug activity. Exp Parasitol 2023; 254:108620. [PMID: 37716462 DOI: 10.1016/j.exppara.2023.108620] [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: 04/28/2023] [Revised: 09/06/2023] [Accepted: 09/12/2023] [Indexed: 09/18/2023]
Abstract
Conditional gene expression is a powerful tool to investigate putative vaccine and drug targets, especially in a haploid organism such as Plasmodium falciparum. Inducible systems based on regulation of either transcription, translation, protein or mRNA stability, among others, allow switching on an off the expression of any desired gene causing specific gain or loss of function phenotypes. However, those systems can be cumbersome involving the construction of large plasmids and generation of multiple transgenic parasite lines. In addition, the dynamic range of regulation achieved is not predictable for each individual gene and can be insufficient to generate detectable phenotypes when the genes of interest are silenced. Here, we combined up to three distinct inducible systems to regulate the expression of a single gene. Expression of the reporter NanoLuc luciferase was regulated over 40-fold, which correlates to the regulation achieved by each individual system multiplied by each other. We applied the conditionally expressed NanoLuc to evaluate the effect of fast-acting antimalarials such as chloroquine and artesunate as well as of slower-acting ones such as atovaquone. The conditionally expressed reporter allowed faster and more reliable detection of toxicity to the parasite, which correlated to the expected action of each compound. Bioluminescence achieved by the expression of this inducible highly sensitive reporter is therefore a promising tool to investigate the temporal effect of potential new antimalarials. This single plasmid combination system might also prove useful to achieve sufficient regulation of genes of interest to produce loss-of-function phenotypes.
Collapse
Affiliation(s)
- Caroline Lima
- Federal University of Sao Paulo, Santos, Sao Paulo, Brazil
| | - Ignasi B Verdaguer
- Departamento de Parasitologia, Instituto de Ciência Biomédicas, Universidade de São Paulo, Avenida Prof. Lineu Prestes, 1374, São Paulo, SP, 05508-900, Brazil
| | - Gerhard Wunderlich
- Departamento de Parasitologia, Instituto de Ciência Biomédicas, Universidade de São Paulo, Avenida Prof. Lineu Prestes, 1374, São Paulo, SP, 05508-900, Brazil
| | - Alejandro M Katzin
- Departamento de Parasitologia, Instituto de Ciência Biomédicas, Universidade de São Paulo, Avenida Prof. Lineu Prestes, 1374, São Paulo, SP, 05508-900, Brazil
| | - Brendan S Crabb
- Burnet Institute, Melbourne, VIC, 3004, Australia; University of Melbourne, VIC, 3052, Australia; Monash University, Melbourne, VIC, 3004, Australia
| | - Paul R Gilson
- Burnet Institute, Melbourne, VIC, 3004, Australia; University of Melbourne, VIC, 3052, Australia
| | | |
Collapse
|
4
|
Xu J, Hu F, Li S, Bao J, Yin Y, Ren Z, Deng Y, Tian F, Bao G, Liu J, Li Y, He X, Xi J, Lu F. Fluorescent Nitrogen-Doped Carbon Dots for Label Live Elder Blood-Stage Plasmodium falciparum through New Permeability Pathways. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27134163. [PMID: 35807422 PMCID: PMC9267939 DOI: 10.3390/molecules27134163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/21/2022] [Accepted: 06/24/2022] [Indexed: 01/11/2023]
Abstract
To verify the size and emergence time of new permeability pathways (NPPs) in malaria parasites, the permeability of the Plasmodium falciparum-infected erythrocytes was tested with different particle sizes of nanomaterials by flow cytometry assay. The results confirmed the permeability of the host cell membrane increases with parasite maturation for the stage-development evolution of NPPs, and especially found that a particle size of about 50 nm had higher efficiency. As a kind of the novel nanomaterials, nitrogen-doped carbon dots (NCDs) showed no toxicity, specificity binding ability to the malaria parasites, and could label live elder blood-stage P. falciparum through NPPs, indicating the potential application in cell imaging. NPPs and some nanomaterials such as NCDs deserve more attention and exploration for the elimination and prevention of malaria.
Collapse
Affiliation(s)
- Jiahui Xu
- Jiangsu Key Laboratory of Experimental and Translational Non-Coding RNA Research, School of Medicine, Yangzhou University, Yangzhou 225009, China; (J.X.); (F.H.); (S.L.); (Y.Y.); (Z.R.); (Y.D.); (F.T.); (Y.L.); (X.H.)
- Affiliated Hospital of Yangzhou University, Yangzhou 225000, China; (J.B.); (G.B.); (J.L.)
| | - Fengyue Hu
- Jiangsu Key Laboratory of Experimental and Translational Non-Coding RNA Research, School of Medicine, Yangzhou University, Yangzhou 225009, China; (J.X.); (F.H.); (S.L.); (Y.Y.); (Z.R.); (Y.D.); (F.T.); (Y.L.); (X.H.)
- The Third People’s Hospital of Yangzhou, Yangzhou 225012, China
| | - Shuang Li
- Jiangsu Key Laboratory of Experimental and Translational Non-Coding RNA Research, School of Medicine, Yangzhou University, Yangzhou 225009, China; (J.X.); (F.H.); (S.L.); (Y.Y.); (Z.R.); (Y.D.); (F.T.); (Y.L.); (X.H.)
| | - Jiaojiao Bao
- Affiliated Hospital of Yangzhou University, Yangzhou 225000, China; (J.B.); (G.B.); (J.L.)
| | - Yi Yin
- Jiangsu Key Laboratory of Experimental and Translational Non-Coding RNA Research, School of Medicine, Yangzhou University, Yangzhou 225009, China; (J.X.); (F.H.); (S.L.); (Y.Y.); (Z.R.); (Y.D.); (F.T.); (Y.L.); (X.H.)
| | - Zhenyu Ren
- Jiangsu Key Laboratory of Experimental and Translational Non-Coding RNA Research, School of Medicine, Yangzhou University, Yangzhou 225009, China; (J.X.); (F.H.); (S.L.); (Y.Y.); (Z.R.); (Y.D.); (F.T.); (Y.L.); (X.H.)
| | - Ying Deng
- Jiangsu Key Laboratory of Experimental and Translational Non-Coding RNA Research, School of Medicine, Yangzhou University, Yangzhou 225009, China; (J.X.); (F.H.); (S.L.); (Y.Y.); (Z.R.); (Y.D.); (F.T.); (Y.L.); (X.H.)
| | - Fang Tian
- Jiangsu Key Laboratory of Experimental and Translational Non-Coding RNA Research, School of Medicine, Yangzhou University, Yangzhou 225009, China; (J.X.); (F.H.); (S.L.); (Y.Y.); (Z.R.); (Y.D.); (F.T.); (Y.L.); (X.H.)
| | - Guangyu Bao
- Affiliated Hospital of Yangzhou University, Yangzhou 225000, China; (J.B.); (G.B.); (J.L.)
| | - Jian Liu
- Affiliated Hospital of Yangzhou University, Yangzhou 225000, China; (J.B.); (G.B.); (J.L.)
| | - Yinyue Li
- Jiangsu Key Laboratory of Experimental and Translational Non-Coding RNA Research, School of Medicine, Yangzhou University, Yangzhou 225009, China; (J.X.); (F.H.); (S.L.); (Y.Y.); (Z.R.); (Y.D.); (F.T.); (Y.L.); (X.H.)
| | - Xinlong He
- Jiangsu Key Laboratory of Experimental and Translational Non-Coding RNA Research, School of Medicine, Yangzhou University, Yangzhou 225009, China; (J.X.); (F.H.); (S.L.); (Y.Y.); (Z.R.); (Y.D.); (F.T.); (Y.L.); (X.H.)
| | - Juqun Xi
- Jiangsu Key Laboratory of Experimental and Translational Non-Coding RNA Research, School of Medicine, Yangzhou University, Yangzhou 225009, China; (J.X.); (F.H.); (S.L.); (Y.Y.); (Z.R.); (Y.D.); (F.T.); (Y.L.); (X.H.)
- Correspondence: (J.X.); (F.L.)
| | - Feng Lu
- Jiangsu Key Laboratory of Experimental and Translational Non-Coding RNA Research, School of Medicine, Yangzhou University, Yangzhou 225009, China; (J.X.); (F.H.); (S.L.); (Y.Y.); (Z.R.); (Y.D.); (F.T.); (Y.L.); (X.H.)
- Affiliated Hospital of Yangzhou University, Yangzhou 225000, China; (J.B.); (G.B.); (J.L.)
- Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Correspondence: (J.X.); (F.L.)
| |
Collapse
|
5
|
Kent RS, Briggs EM, Colon BL, Alvarez C, Silva Pereira S, De Niz M. Paving the Way: Contributions of Big Data to Apicomplexan and Kinetoplastid Research. Front Cell Infect Microbiol 2022; 12:900878. [PMID: 35734575 PMCID: PMC9207352 DOI: 10.3389/fcimb.2022.900878] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/06/2022] [Indexed: 11/13/2022] Open
Abstract
In the age of big data an important question is how to ensure we make the most out of the resources we generate. In this review, we discuss the major methods used in Apicomplexan and Kinetoplastid research to produce big datasets and advance our understanding of Plasmodium, Toxoplasma, Cryptosporidium, Trypanosoma and Leishmania biology. We debate the benefits and limitations of the current technologies, and propose future advancements that may be key to improving our use of these techniques. Finally, we consider the difficulties the field faces when trying to make the most of the abundance of data that has already been, and will continue to be, generated.
Collapse
Affiliation(s)
- Robyn S. Kent
- Department of Microbiology and Molecular Genetics, University of Vermont, Burlington, VT, United States
| | - Emma M. Briggs
- Institute for Immunology and Infection Research, School of Biological Sciences, University Edinburgh, Edinburgh, United Kingdom
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Beatrice L. Colon
- Wellcome Centre for Anti-Infectives Research, Division of Biological Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Catalina Alvarez
- de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Sara Silva Pereira
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | - Mariana De Niz
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
- Institut Pasteur, Paris, France
| |
Collapse
|
6
|
Ullah I, Sharma R, Mete A, Biagini GA, Wetzel DM, Horrocks PD. The relative rate of kill of the MMV Malaria Box compounds provides links to the mode of antimalarial action and highlights scaffolds of medicinal chemistry interest. J Antimicrob Chemother 2021; 75:362-370. [PMID: 31665424 DOI: 10.1093/jac/dkz443] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 09/04/2019] [Accepted: 10/01/2019] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES Rapid rate-of-kill (RoK) is a key parameter in the target candidate profile 1 (TCP1) for the next-generation antimalarial drugs for uncomplicated malaria, termed Single Encounter Radical Cure and Prophylaxis (SERCaP). TCP1 aims to rapidly eliminate the initial parasite burden, ideally as fast as artesunate, but minimally as fast as chloroquine. Here we explore whether the relative RoK of the Medicine for Malaria Venture (MMV) Malaria Box compounds is linked to their mode of action (MoA) and identify scaffolds of medicinal chemistry interest. METHODS We used a bioluminescence relative RoK (BRRoK) assay over 6 and 48 h, with exposure to equipotent IC50 concentrations, to compare the cytocidal effects of Malaria Box compounds with those of benchmark antimalarials. RESULTS BRRoK assay data demonstrate the following relative RoKs, from fast to slow: inhibitors of PfATP4>parasite haemoglobin catabolism>dihydrofolate reductase-thymidylate synthase (DHFR-TS)>dihydroorotate dehydrogenase (DHODH)>bc1 complex. Core-scaffold clustering analyses revealed intrinsic rapid cytocidal action for diamino-glycerols and 2-(aminomethyl)phenol, but slow action for 2-phenylbenz-imidazoles, 8-hydroxyquinolines and triazolopyrimidines. CONCLUSIONS This study provides proof of principle that a compound's RoK is related to its MoA and that the target's intrinsic RoK is also modified by factors affecting a drug's access to it. Our findings highlight that as we use medicinal chemistry to improve potency, we can also improve the RoK for some scaffolds. Our BRRoK assay provides the necessary throughput for drug discovery and a critical decision-making tool to support development campaigns. Finally, two scaffolds, diamino-glycerols and 2-phenylbenzimidazoles, exhibit fast cytocidal action, inviting medicinal chemistry improvements towards TCP1 candidates.
Collapse
Affiliation(s)
- Imran Ullah
- Institute for Science and Technology in Medicine, Keele University, Staffordshire, UK
| | - Raman Sharma
- Research Centre for Drugs and Diagnostics, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, UK
| | - Antonio Mete
- Medsyndesign Ltd, Advanced Technology Innovation Centre, 5 Oakwood Drive, Loughborough, UK
| | - Giancarlo A Biagini
- Research Centre for Drugs and Diagnostics, Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, UK
| | - Dawn M Wetzel
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
| | - Paul D Horrocks
- Institute for Science and Technology in Medicine, Keele University, Staffordshire, UK
| |
Collapse
|
7
|
Hameed H, King EFB, Doleckova K, Bartholomew B, Hollinshead J, Mbye H, Ullah I, Walker K, Van Veelen M, Abou-Akkada SS, Nash RJ, Horrocks PD, Price HP. Temperate Zone Plant Natural Products-A Novel Resource for Activity against Tropical Parasitic Diseases. Pharmaceuticals (Basel) 2021; 14:227. [PMID: 33800005 PMCID: PMC7998250 DOI: 10.3390/ph14030227] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/22/2021] [Accepted: 03/03/2021] [Indexed: 11/16/2022] Open
Abstract
The use of plant-derived natural products for the treatment of tropical parasitic diseases often has ethnopharmacological origins. As such, plants grown in temperate regions remain largely untested for novel anti-parasitic activities. We describe here a screen of the PhytoQuest Phytopure library, a novel source comprising over 600 purified compounds from temperate zone plants, against in vitro culture systems for Plasmodium falciparum, Leishmania mexicana, Trypanosoma evansi and T. brucei. Initial screen revealed 6, 65, 15 and 18 compounds, respectively, that decreased each parasite's growth by at least 50% at 1-2 µM concentration. These initial hits were validated in concentration-response assays against the parasite and the human HepG2 cell line, identifying hits with EC50 < 1 μM and a selectivity index of >10. Two sesquiterpene glycosides were identified against P. falciparum, four sterols against L. mexicana, and five compounds of various scaffolds against T. brucei and T. evansi. An L. mexicana resistant line was generated for the sterol 700022, which was found to have cross-resistance to the anti-leishmanial drug miltefosine as well as to the other leishmanicidal sterols. This study highlights the potential of a temperate plant secondary metabolites as a novel source of natural products against tropical parasitic diseases.
Collapse
Affiliation(s)
- Hamza Hameed
- Centre for Applied Entomology and Parasitology, Keele University, Staffordshire ST5 5BG, UK; (H.H.); (E.F.B.K.); (K.D.); (H.M.); (I.U.); (M.V.V.)
- Department of Chemistry, College of Education for Pure Science, University of Mosul, Mosul, Iraq
| | - Elizabeth F. B. King
- Centre for Applied Entomology and Parasitology, Keele University, Staffordshire ST5 5BG, UK; (H.H.); (E.F.B.K.); (K.D.); (H.M.); (I.U.); (M.V.V.)
| | - Katerina Doleckova
- Centre for Applied Entomology and Parasitology, Keele University, Staffordshire ST5 5BG, UK; (H.H.); (E.F.B.K.); (K.D.); (H.M.); (I.U.); (M.V.V.)
- Department of Biology, Faculty of Life Sciences, University of Hradec Králové, 500 03 Hradec Králové, Czech Republic
| | | | | | - Haddijatou Mbye
- Centre for Applied Entomology and Parasitology, Keele University, Staffordshire ST5 5BG, UK; (H.H.); (E.F.B.K.); (K.D.); (H.M.); (I.U.); (M.V.V.)
- MRC Unit The Gambia at LSHTM, Atlantic Boulevard, Fajara, Banjul PO Box 273, The Gambia
| | - Imran Ullah
- Centre for Applied Entomology and Parasitology, Keele University, Staffordshire ST5 5BG, UK; (H.H.); (E.F.B.K.); (K.D.); (H.M.); (I.U.); (M.V.V.)
- Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA 02115, USA
| | - Karen Walker
- School of Life Sciences, Keele University, Staffordshire ST5 5BG, UK;
| | - Maria Van Veelen
- Centre for Applied Entomology and Parasitology, Keele University, Staffordshire ST5 5BG, UK; (H.H.); (E.F.B.K.); (K.D.); (H.M.); (I.U.); (M.V.V.)
| | | | - Robert J. Nash
- PhytoQuest Limited, Aberystwyth SY23 3EB, UK; (B.B.); (J.H.); (R.J.N.)
| | - Paul D. Horrocks
- Centre for Applied Entomology and Parasitology, Keele University, Staffordshire ST5 5BG, UK; (H.H.); (E.F.B.K.); (K.D.); (H.M.); (I.U.); (M.V.V.)
| | - Helen P. Price
- Centre for Applied Entomology and Parasitology, Keele University, Staffordshire ST5 5BG, UK; (H.H.); (E.F.B.K.); (K.D.); (H.M.); (I.U.); (M.V.V.)
| |
Collapse
|
8
|
Lawong A, Gahalawat S, Okombo J, Striepen J, Yeo T, Mok S, Deni I, Bridgford JL, Niederstrasser H, Zhou A, Posner B, Wittlin S, Gamo FJ, Crespo B, Churchyard A, Baum J, Mittal N, Winzeler E, Laleu B, Palmer MJ, Charman SA, Fidock DA, Ready JM, Phillips MA. Novel Antimalarial Tetrazoles and Amides Active against the Hemoglobin Degradation Pathway in Plasmodium falciparum. J Med Chem 2021; 64:2739-2761. [PMID: 33620219 DOI: 10.1021/acs.jmedchem.0c02022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Malaria control programs continue to be threatened by drug resistance. To identify new antimalarials, we conducted a phenotypic screen and identified a novel tetrazole-based series that shows fast-kill kinetics and a relatively low propensity to develop high-level resistance. Preliminary structure-activity relationships were established including identification of a subseries of related amides with antiplasmodial activity. Assaying parasites with resistance to antimalarials led us to test whether the series had a similar mechanism of action to chloroquine (CQ). Treatment of synchronized Plasmodium falciparum parasites with active analogues revealed a pattern of intracellular inhibition of hemozoin (Hz) formation reminiscent of CQ's action. Drug selections yielded only modest resistance that was associated with amplification of the multidrug resistance gene 1 (pfmdr1). Thus, we have identified a novel chemical series that targets the historically druggable heme polymerization pathway and that can form the basis of future optimization efforts to develop a new malaria treatment.
Collapse
Affiliation(s)
- Aloysus Lawong
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Suraksha Gahalawat
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - John Okombo
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, New York 10032, United States
| | - Josefine Striepen
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, New York 10032, United States
| | - Tomas Yeo
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, New York 10032, United States
| | - Sachel Mok
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, New York 10032, United States
| | - Ioanna Deni
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, New York 10032, United States
| | - Jessica L Bridgford
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, New York 10032, United States
| | - Hanspeter Niederstrasser
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Anwu Zhou
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Bruce Posner
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Sergio Wittlin
- Swiss Tropical and Public Health Institute, 4002 Basel, Switzerland.,University of Basel, 4002 Basel, Switzerland
| | | | - Benigno Crespo
- Medicines Development Campus, GlaxoSmithKline, Tres Cantos, 28760 Madrid, Spain
| | - Alisje Churchyard
- Department of Life Sciences, Imperial College London, SW7 2AZ South Kensington, U.K
| | - Jake Baum
- Department of Life Sciences, Imperial College London, SW7 2AZ South Kensington, U.K
| | - Nimisha Mittal
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, California 92093, United States
| | - Elizabeth Winzeler
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, California 92093, United States
| | - Benoît Laleu
- Medicines for Malaria Venture, 1215 Geneva, Switzerland
| | | | - Susan A Charman
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - David A Fidock
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, New York 10032, United States.,Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, New York 10032, United States
| | - Joseph M Ready
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Margaret A Phillips
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| |
Collapse
|
9
|
Rapid and quantitative antimalarial drug efficacy testing via the magneto-optical detection of hemozoin. Sci Rep 2020; 10:14025. [PMID: 32820190 PMCID: PMC7441145 DOI: 10.1038/s41598-020-70860-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 07/24/2020] [Indexed: 01/24/2023] Open
Abstract
Emergence of resistant Plasmodium species makes drug efficacy testing a crucial part of malaria control. Here we describe a novel assay for sensitive, fast and simple drug screening via the magneto-optical detection of hemozoin, a natural biomarker formed during the hemoglobin metabolism of Plasmodium species. By quantifying hemozoin production over the intraerythrocytic cycle, we reveal that hemozoin formation is already initiated by ~ 6–12 h old ring-stage parasites. We demonstrate that the new assay is capable of drug efficacy testing with incubation times as short as 6–10 h, using synchronized P. falciparum 3D7 cultures incubated with chloroquine, piperaquine and dihydroartemisinin. The determined 50% inhibitory concentrations agree well with values established by standard assays requiring significantly longer testing time. Accordingly, we conclude that magneto-optical hemozoin detection provides a practical approach for the quick assessment of drug effect with short incubation times, which may also facilitate stage-specific assessment of drug inhibitory effects.
Collapse
|
10
|
Ullah I, Gahalawat S, Booshehri LM, Niederstrasser H, Majumdar S, Leija C, Bradford JM, Hu B, Ready JM, Wetzel DM. An Antiparasitic Compound from the Medicines for Malaria Venture Pathogen Box Promotes Leishmania Tubulin Polymerization. ACS Infect Dis 2020; 6:2057-2072. [PMID: 32686409 PMCID: PMC8059355 DOI: 10.1021/acsinfecdis.0c00122] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The few frontline antileishmanial drugs are poorly effective and toxic. To search for new drugs for this neglected tropical disease, we tested the activity of compounds in the Medicines for Malaria Venture (MMV) "Pathogen Box" against Leishmania amazonensis axenic amastigotes. Screening yielded six discovery antileishmanial compounds with EC50 values from 50 to 480 nM. Concentration-response assays demonstrated that the best hit, MMV676477, had mid-nanomolar cytocidal potency against intracellular Leishmania amastigotes, Trypanosoma brucei, and Plasmodium falciparum, suggesting broad antiparasitic activity. We explored structure-activity relationships (SAR) within a small group of MMV676477 analogs and observed a wide potency range (20-5000 nM) against axenic Leishmania amastigotes. Compared to MMV676477, our most potent analog, SW41, had ∼5-fold improved antileishmanial potency. Multiple lines of evidence suggest that MMV676477 selectively disrupts Leishmania tubulin dynamics. Morphological studies indicated that MMV676477 and analogs affected L. amazonensis during cell division. Differential centrifugation showed that MMV676477 promoted partitioning of cellular tubulin toward the polymeric form in parasites. Turbidity assays with purified Leishmania and porcine tubulin demonstrated that MMV676477 promoted leishmanial tubulin polymerization in a concentration-dependent manner. Analogs' antiparasitic activity correlated with their ability to facilitate purified Leishmania tubulin polymerization. Chemical cross-linking demonstrated binding of the MMV676477 scaffold to purified Leishmania tubulin, and competition studies established a correlation between binding and antileishmanial activity. Our studies demonstrate that MMV676477 is a potent antiparasitic compound that preferentially promotes Leishmania microtubule polymerization. Due to its selectivity for and broad-spectrum activity against multiple parasites, this scaffold shows promise for antiparasitic drug development.
Collapse
Affiliation(s)
- Imran Ullah
- Department of Pediatrics and Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Suraksha Gahalawat
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Laela M. Booshehri
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Hanspeter Niederstrasser
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Shreoshi Majumdar
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Christopher Leija
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - James M. Bradford
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Bin Hu
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Joseph M. Ready
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Dawn M. Wetzel
- Department of Pediatrics and Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| |
Collapse
|
11
|
Czechowski T, Rinaldi MA, Famodimu MT, Van Veelen M, Larson TR, Winzer T, Rathbone DA, Harvey D, Horrocks P, Graham IA. Flavonoid Versus Artemisinin Anti-malarial Activity in Artemisia annua Whole-Leaf Extracts. FRONTIERS IN PLANT SCIENCE 2019; 10:984. [PMID: 31417596 PMCID: PMC6683762 DOI: 10.3389/fpls.2019.00984] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 07/12/2019] [Indexed: 05/05/2023]
Abstract
Artemisinin, a sesquiterpene lactone produced by Artemisia annua glandular secretory trichomes, is the active ingredient in the most effective treatment for uncomplicated malaria caused by Plasmodium falciparum parasites. Other metabolites in A. annua or related species, particularly flavonoids, have been proposed to either act as antimalarials on their own or act synergistically with artemisinin to enhance antimalarial activity. We identified a mutation that disrupts the CHALCONE ISOMERASE 1 (CHI1) enzyme that is responsible for the second committed step of flavonoid biosynthesis. Detailed metabolite profiling revealed that chi1-1 lacks all major flavonoids but produces wild-type artemisinin levels, making this mutant a useful tool to test the antiplasmodial effects of flavonoids. We used whole-leaf extracts from chi1-1 and mutant lines impaired in artemisinin production in bioactivity in vitro assays against intraerythrocytic P. falciparum Dd2. We found that chi1-1 extracts did not differ from wild-type extracts in antiplasmodial efficacy nor initial rate of cytocidal action. Furthermore, extracts from the A. annua cyp71av1-1 mutant and RNAi lines impaired in amorpha-4,11-diene synthase gene expression, which are both severely compromised in artemisinin biosynthesis but unaffected in flavonoid metabolism, showed very low or no antiplasmodial activity. These results demonstrate that in vitro bioactivity against P. falciparum of flavonoids is negligible when compared to that of artemisinin.
Collapse
Affiliation(s)
- Tomasz Czechowski
- Centre for Novel Agricultural Products, Department of Biology, University of York, York, United Kingdom
| | - Mauro A. Rinaldi
- Centre for Novel Agricultural Products, Department of Biology, University of York, York, United Kingdom
| | | | | | - Tony R. Larson
- Centre for Novel Agricultural Products, Department of Biology, University of York, York, United Kingdom
| | - Thilo Winzer
- Centre for Novel Agricultural Products, Department of Biology, University of York, York, United Kingdom
| | - Deborah A. Rathbone
- Centre for Novel Agricultural Products, Department of Biology, University of York, York, United Kingdom
- Biorenewables Development Centre, Dunnington, United Kingdom
| | - David Harvey
- Centre for Novel Agricultural Products, Department of Biology, University of York, York, United Kingdom
| | - Paul Horrocks
- Institute for Science and Technology in Medicine, Keele University, Keele, United Kingdom
- School of Medicine, Keele University, Keele, United Kingdom
| | - Ian A. Graham
- Centre for Novel Agricultural Products, Department of Biology, University of York, York, United Kingdom
- *Correspondence: Ian A. Graham,
| |
Collapse
|
12
|
Berry SL, Hameed H, Thomason A, Maciej-Hulme ML, Saif Abou-Akkada S, Horrocks P, Price HP. Development of NanoLuc-PEST expressing Leishmania mexicana as a new drug discovery tool for axenic- and intramacrophage-based assays. PLoS Negl Trop Dis 2018; 12:e0006639. [PMID: 30001317 PMCID: PMC6057649 DOI: 10.1371/journal.pntd.0006639] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 07/24/2018] [Accepted: 06/26/2018] [Indexed: 12/12/2022] Open
Abstract
The protozoan parasite Leishmania causes leishmaniasis; a spectrum of diseases of which there are an estimated 1 million new cases each year. Current treatments are toxic, expensive, difficult to administer, and resistance to them is emerging. New therapeutics are urgently needed, however, screening the infective amastigote form of the parasite is challenging. Only certain species can be differentiated into axenic amastigotes, and compound activity against these does not always correlate with efficacy against the parasite in its intracellular niche. Methods used to assess compound efficacy on intracellular amastigotes often rely on microscopy-based assays. These are laborious, require specialist equipment and can only determine parasite burden, not parasite viability. We have addressed this clear need in the anti-leishmanial drug discovery process by producing a transgenic L. mexicana cell line that expresses the luciferase NanoLuc-PEST. We tested the sensitivity and versatility of this transgenic strain, in comparison with strains expressing NanoLuc and the red-shifted firefly luciferase. We then compared the NanoLuc-PEST luciferase to the current methods in both axenic and intramacrophage amastigotes following treatment with a supralethal dose of Amphotericin B. NanoLuc-PEST was a more dynamic indicator of cell viability due to its high turnover rate and high signal:background ratio. This, coupled with its sensitivity in the intramacrophage assay, led us to validate the NanoLuc-PEST expressing cell line using the MMV Pathogen Box in a two-step process: i) identify hits against axenic amastigotes, ii) screen these hits using our bioluminescence-based intramacrophage assay. The data obtained from this highlights the potential of compounds active against M. tuberculosis to be re-purposed for use against Leishmania. Our transgenic L. mexicana cell line is therefore a highly sensitive and dynamic system suitable for Leishmania drug discovery in axenic and intramacrophage amastigote models. The protozoan parasite Leishmania causes a spectrum of diseases collectively known as leishmaniasis. The parasite is transmitted to humans by the bite of its vector, the sand fly, following which the parasite invades host white blood cells, particularly macrophages. Leishmaniasis is classified as a neglected tropical disease, and is endemic in 97 countries. Symptoms of the disease depend on the species of Leishmania. These include skin lesions, destruction of the mucosal membranes, and the visceral form which is usually fatal if untreated. Current therapeutic options for leishmaniasis have a number of associated problems that include toxicity, the development of drug resistance and poor patient compliance due to lengthy and painful treatment regimens. New therapeutics are therefore urgently needed. The ability to screen potential drug candidates requires robust screening assays. Currently, screening the intracellular parasite relies on microscopy-based techniques that require expensive equipment, are time consuming and only detect parasite burden, not viability. By using a transgenic cell line that expresses the NanoLuc-PEST luciferase, we show that we have a parasite-specific viability marker that can be used to measure the efficacy of compounds against the intracellular parasite. We validate the potential of this cell line by screening the MMV Pathogen Box.
Collapse
Affiliation(s)
- Sarah L. Berry
- Centre for Applied Entomology and Parasitology, School of Life Sciences, Keele University, Newcastle-under-Lyme, Staffordshire, United Kingdom
| | - Hamza Hameed
- Institute for Science and Technology in Medicine, Keele University, Newcastle-under-Lyme, Staffordshire, United Kingdom
| | - Anna Thomason
- Centre for Applied Entomology and Parasitology, School of Life Sciences, Keele University, Newcastle-under-Lyme, Staffordshire, United Kingdom
- Current address: School of Environment and Life Sciences, University of Salford, Salford, United Kingdom
| | - Marissa L. Maciej-Hulme
- Radboud University Medical Center, Department of Nephrology, Geert Grooteplein 10, GA Nijmegan, The Netherlands
| | - Somaia Saif Abou-Akkada
- Centre for Applied Entomology and Parasitology, School of Life Sciences, Keele University, Newcastle-under-Lyme, Staffordshire, United Kingdom
- Faculty of Veterinary Medicine, Alexandria University, Alexandria, Egypt
| | - Paul Horrocks
- Centre for Applied Entomology and Parasitology, School of Life Sciences, Keele University, Newcastle-under-Lyme, Staffordshire, United Kingdom
- Institute for Science and Technology in Medicine, Keele University, Newcastle-under-Lyme, Staffordshire, United Kingdom
| | - Helen P. Price
- Centre for Applied Entomology and Parasitology, School of Life Sciences, Keele University, Newcastle-under-Lyme, Staffordshire, United Kingdom
- * E-mail:
| |
Collapse
|
13
|
G-Quadruplex DNA Motifs in the Malaria Parasite Plasmodium falciparum and Their Potential as Novel Antimalarial Drug Targets. Antimicrob Agents Chemother 2018; 62:AAC.01828-17. [PMID: 29311059 DOI: 10.1128/aac.01828-17] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 12/20/2017] [Indexed: 01/06/2023] Open
Abstract
G-quadruplexes are DNA or RNA secondary structures that can be formed from guanine-rich nucleic acids. These four-stranded structures, composed of stacked quartets of guanine bases, can be highly stable and have been demonstrated to occur in vivo in the DNA of human cells and other systems, where they play important biological roles, influencing processes such as telomere maintenance, DNA replication and transcription, or, in the case of RNA G-quadruplexes, RNA translation and processing. We report for the first time that DNA G-quadruplexes can be detected in the nuclei of the malaria parasite Plasmodium falciparum, which has one of the most A/T-biased genomes sequenced and therefore possesses few guanine-rich sequences with the potential to form G-quadruplexes. We show that despite this paucity of putative G-quadruplex-forming sequences, P. falciparum parasites are sensitive to several G-quadruplex-stabilizing drugs, including quarfloxin, which previously reached phase 2 clinical trials as an anticancer drug. Quarfloxin has a rapid initial rate of kill and is active against ring stages as well as replicative stages of intraerythrocytic development. We show that several G-quadruplex-stabilizing drugs, including quarfloxin, can suppress the transcription of a G-quadruplex-containing reporter gene in P. falciparum but that quarfloxin does not appear to disrupt the transcription of rRNAs, which was proposed as its mode of action in both human cells and trypanosomes. These data suggest that quarfloxin has potential for repositioning as an antimalarial with a novel mode of action. Furthermore, G-quadruplex biology in P. falciparum may present a target for development of other new antimalarial drugs.
Collapse
|
14
|
Gisselberg JE, Herrera Z, Orchard LM, Llinás M, Yeh E. Specific Inhibition of the Bifunctional Farnesyl/Geranylgeranyl Diphosphate Synthase in Malaria Parasites via a New Small-Molecule Binding Site. Cell Chem Biol 2017; 25:185-193.e5. [PMID: 29276048 DOI: 10.1016/j.chembiol.2017.11.010] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 09/24/2017] [Accepted: 11/22/2017] [Indexed: 12/19/2022]
Abstract
The bifunctional farnesyl/geranylgeranyl diphosphate synthase (FPPS/GGPPS) is a key branchpoint enzyme in isoprenoid biosynthesis in Plasmodium falciparum (malaria) parasites. PfFPPS/GGPPS is a validated, high-priority antimalarial drug target. Unfortunately, current bisphosphonate drugs that inhibit FPPS and GGPPS enzymes by acting as a diphosphate substrate analog show poor bioavailability and selectivity for PfFPPS/GGPPS. We identified a new non-bisphosphonate compound, MMV019313, which is highly selective for PfFPPS/GGPPS and showed no activity against human FPPS or GGPPS. Inhibition of PfFPPS/GGPPS by MMV019313, but not bisphosphonates, was disrupted in an S228T variant, demonstrating that MMV019313 and bisphosphonates have distinct modes of inhibition. Molecular docking indicated that MMV019313 did not bind previously characterized substrate sites in PfFPPS/GGPPS. Our finding uncovers a new, selective small-molecule binding site in this important antimalarial drug target with superior druggability compared with the known inhibitor site and sets the stage for the development of Plasmodium-specific FPPS/GGPPS inhibitors.
Collapse
Affiliation(s)
- Jolyn E Gisselberg
- Department of Biochemistry, Stanford Medical School, Stanford University, Stanford, CA 94305, USA
| | - Zachary Herrera
- Department of Biochemistry, Stanford Medical School, Stanford University, Stanford, CA 94305, USA
| | - Lindsey M Orchard
- Department of Biochemistry & Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA; Huck Center for Malaria Research, Pennsylvania State University, University Park, PA 16802, USA
| | - Manuel Llinás
- Department of Biochemistry & Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA; Huck Center for Malaria Research, Pennsylvania State University, University Park, PA 16802, USA; Department of Chemistry, Pennsylvania State University, University Park, PA 16802, USA
| | - Ellen Yeh
- Department of Biochemistry, Stanford Medical School, Stanford University, Stanford, CA 94305, USA; Department of Pathology, Stanford Medical School, Stanford University, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford Medical School, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|