1
|
Garnie LF, Egan TJ, Wicht KJ. Heme Detoxification in the Malaria Parasite Plasmodium falciparum: A Time-Dependent Basal-Level Analysis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.06.641703. [PMID: 40093040 PMCID: PMC11908274 DOI: 10.1101/2025.03.06.641703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Malaria is a deadly disease for which therapeutic options are threatened by the rise of antimalarial resistance. Inhibiting the formation of hemozoin (the product of heme detoxification) in the digestive vacuole (DV) is the mechanism of action of numerous antimalarial drugs, including those in development as new therapies. This drug target remains attractive as hemozoin is an abiotic and non-mutable molecule, unique to the parasite. The underlying parasite biology of the heme detoxification pathway is complex and requires a deeper understanding. This study focuses on the DV of Plasmodium falciparum, utilizing confocal microscopy, immunoblotting and cellular fractionation techniques to study its native state over time. Using parameters such as the uptake into and growth of the DV, relative abundance of plasmepsins (PMs) I and IV and basal levels of hemoglobin, heme and hemozoin, it was found that DV physiology in chloroquine (CQ)-sensitive NF54 parasites follows three distinct developmental phases: the lag-type growth (20 to 28 h), rapid growth phase (28 to 40 h) and the plateau (40 to 48 h). These phases hold specific characteristics with respect to the investigated parameters. In addition, key differences between CQ-sensitive NF54 and CQ-resistant Dd2 parasites were observed.
Collapse
Affiliation(s)
- Larnelle F Garnie
- Department of Chemistry, Faculty of Science, University of Cape Town, Rondebosch 7701, South Africa
| | - Timothy J Egan
- Department of Chemistry, Faculty of Science, University of Cape Town, Rondebosch 7701, South Africa
- Institute of Infectious Diseases and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
| | - Kathryn J Wicht
- Department of Chemistry, Faculty of Science, University of Cape Town, Rondebosch 7701, South Africa
- Institute of Infectious Diseases and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
- Holistic Drug Discovery and Development (H3D) Center, University of Cape Town, Rondebosch 7701, South Africa
| |
Collapse
|
2
|
Luth MR, Godinez-Macias KP, Chen D, Okombo J, Thathy V, Cheng X, Daggupati S, Davies H, Dhingra SK, Economy JM, Edgar RCS, Gomez-Lorenzo MG, Istvan ES, Jado JC, LaMonte GM, Melillo B, Mok S, Narwal SK, Ndiaye T, Ottilie S, Diaz SP, Park H, Peña S, Rocamora F, Sakata-Kato T, Small-Saunders JL, Summers RL, Tumwebaze PK, Vanaerschot M, Xia G, Yeo T, You A, Gamo FJ, Goldberg DE, Lee MC, McNamara CW, Ndiaye D, Rosenthal PJ, Schreiber SL, Serra G, De Siqueira-Neto JL, Skinner-Adams TS, Uhlemann AC, Kato N, Lukens AK, Wirth DF, Fidock DA, Winzeler EA. Systematic in vitro evolution in Plasmodium falciparum reveals key determinants of drug resistance. Science 2024; 386:eadk9893. [PMID: 39607932 PMCID: PMC11809290 DOI: 10.1126/science.adk9893] [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: 10/02/2023] [Revised: 05/21/2024] [Accepted: 10/07/2024] [Indexed: 11/30/2024]
Abstract
Surveillance of drug resistance and the discovery of novel targets-key objectives in the fight against malaria-rely on identifying resistance-conferring mutations in Plasmodium parasites. Current approaches, while successful, require laborious experimentation or large sample sizes. To elucidate shared determinants of antimalarial resistance that can empower in silico inference, we examined the genomes of 724 Plasmodium falciparum clones, each selected in vitro for resistance to one of 118 compounds. We identified 1448 variants in 128 recurrently mutated genes, including drivers of antimalarial multidrug resistance. In contrast to naturally occurring variants, those selected in vitro are more likely to be missense or frameshift, involve bulky substitutions, and occur in conserved, ordered protein domains. Collectively, our dataset reveals mutation features that predict drug resistance in eukaryotic pathogens.
Collapse
Affiliation(s)
- Madeline R. Luth
- Department of Pediatrics, University of California San Diego; La Jolla, CA 92093, USA
| | | | - Daisy Chen
- Department of Pediatrics, University of California San Diego; La Jolla, CA 92093, USA
| | - John Okombo
- Department of Microbiology and Immunology, Columbia University Irving Medical Center; New York, New York 10032, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center; New York, New York 10032, USA
| | - Vandana Thathy
- Department of Microbiology and Immunology, Columbia University Irving Medical Center; New York, New York 10032, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center; New York, New York 10032, USA
| | - Xiu Cheng
- Global Health Drug Discovery Institute; Beijing, 100192, China
| | - Sindhu Daggupati
- Department of Pediatrics, University of California San Diego; La Jolla, CA 92093, USA
| | - Heledd Davies
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, United Kingdom
| | - Satish K. Dhingra
- Department of Microbiology and Immunology, Columbia University Irving Medical Center; New York, New York 10032, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center; New York, New York 10032, USA
| | - Jan M. Economy
- Department of Pediatrics, University of California San Diego; La Jolla, CA 92093, USA
| | - Rebecca C. S. Edgar
- Biological Chemistry and Drug Discovery, Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, DD1 5EH, United Kingdom
| | | | - Eva S. Istvan
- Department of Internal Medicine, Division of Infectious Diseases, Washington University School of Medicine; Saint Louis, MO 63130, USA
- Department of Molecular Microbiology, Washington University School of Medicine; Saint Louis, MO 63130, USA
| | - Juan Carlos Jado
- Department of Pediatrics, University of California San Diego; La Jolla, CA 92093, USA
| | - Gregory M. LaMonte
- Department of Pediatrics, University of California San Diego; La Jolla, CA 92093, USA
| | - Bruno Melillo
- Chemical Biology and Therapeutics Science Program, Broad Institute; Cambridge, MA 02142, USA
| | - Sachel Mok
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center; New York, New York 10032, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center; New York, New York 10032, USA
| | - Sunil K. Narwal
- Department of Microbiology and Immunology, Columbia University Irving Medical Center; New York, New York 10032, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center; New York, New York 10032, USA
| | - Tolla Ndiaye
- Department of Microbiology and Immunology, Columbia University Irving Medical Center; New York, New York 10032, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center; New York, New York 10032, USA
| | - Sabine Ottilie
- Department of Pediatrics, University of California San Diego; La Jolla, CA 92093, USA
| | - Sara Palomo Diaz
- Global Health Medicines R&D, GSK; Tres Cantos, Madrid 28760, Spain
| | - Heekuk Park
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center; New York, New York 10032, USA
| | - Stella Peña
- Química Farmacéutica, Departamento de Química Orgánica, Facultad de Química, Universidad de la República; Montevideo, Montevideo CC1157, Uruguay
| | - Frances Rocamora
- Department of Pediatrics, University of California San Diego; La Jolla, CA 92093, USA
| | - Tomoyo Sakata-Kato
- Global Health Drug Discovery Institute; Beijing, 100192, China
- Department of Protozoology, Nekken Institute for Tropical Medicine, Nagasaki University; Nagasaki, 852-8523, Japan
| | - Jennifer L. Small-Saunders
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center; New York, New York 10032, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center; New York, New York 10032, USA
| | - Robert L. Summers
- Department of Immunology & Infectious Diseases, Harvard T.H. Chan School of Public Health; Boston, MA 02115, USA
- Infectious Disease and Microbiome Program, The Broad Institute; Cambridge, MA 02142, USA
| | | | - Manu Vanaerschot
- Department of Microbiology and Immunology, Columbia University Irving Medical Center; New York, New York 10032, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center; New York, New York 10032, USA
| | - Guoqin Xia
- Department of Chemistry, The Scripps Research Institute; La Jolla, CA 92037, USA
| | - Tomas Yeo
- Department of Microbiology and Immunology, Columbia University Irving Medical Center; New York, New York 10032, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center; New York, New York 10032, USA
| | - Ashley You
- Department of Pediatrics, University of California San Diego; La Jolla, CA 92093, USA
| | | | - Daniel E. Goldberg
- Department of Internal Medicine, Division of Infectious Diseases, Washington University School of Medicine; Saint Louis, MO 63130, USA
- Department of Molecular Microbiology, Washington University School of Medicine; Saint Louis, MO 63130, USA
| | - Marcus C.S. Lee
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, CB10 1SA, United Kingdom
- Biological Chemistry and Drug Discovery, Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, DD1 5EH, United Kingdom
| | - Case W. McNamara
- Calibr, a division of The Scripps Research Institute; La Jolla, CA 92037, USA
| | - Daouda Ndiaye
- Centre International de Recherche et de Formation en Génomique Appliquée et de Surveillance Sanitaire (CIGASS), Dakar, Senegal
| | - Philip J. Rosenthal
- Department of Medicine, University of California San Francisco; San Francisco, CA 94115, USA
| | | | - Gloria Serra
- Química Farmacéutica, Departamento de Química Orgánica, Facultad de Química, Universidad de la República; Montevideo, Montevideo CC1157, Uruguay
| | - Jair Lage De Siqueira-Neto
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego; La Jolla, CA 92037, USA
| | - Tina S. Skinner-Adams
- Griffith Institute for Drug Discovery, Griffith University; Nathan, Queensland 4111, Australia
| | - Anne-Catrin Uhlemann
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center; New York, New York 10032, USA
| | - Nobutaka Kato
- Global Health Drug Discovery Institute; Beijing, 100192, China
- Department of Protozoology, Nekken Institute for Tropical Medicine, Nagasaki University; Nagasaki, 852-8523, Japan
| | - Amanda K. Lukens
- Department of Immunology & Infectious Diseases, Harvard T.H. Chan School of Public Health; Boston, MA 02115, USA
- Infectious Disease and Microbiome Program, The Broad Institute; Cambridge, MA 02142, USA
| | - Dyann F. Wirth
- Department of Immunology & Infectious Diseases, Harvard T.H. Chan School of Public Health; Boston, MA 02115, USA
- Infectious Disease and Microbiome Program, The Broad Institute; Cambridge, MA 02142, USA
| | - David A. Fidock
- Department of Microbiology and Immunology, Columbia University Irving Medical Center; New York, New York 10032, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center; New York, New York 10032, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center; New York, New York 10032, USA
| | - Elizabeth A. Winzeler
- Department of Pediatrics, University of California San Diego; La Jolla, CA 92093, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego; La Jolla, CA 92037, USA
| |
Collapse
|
3
|
Oyeneyin OE, Moodley R, Mashaba C, Garnie LF, Omoboyowa DA, Rakodi GH, Maphoru MV, Balogun MO, Hoppe HC, Egan TJ, Tukulula M. In vitro antiplasmodium and antitrypanosomal activities, β-haematin formation inhibition, molecular docking and DFT computational studies of quinoline-urea-benzothiazole hybrids. Heliyon 2024; 10:e38434. [PMID: 39397937 PMCID: PMC11471183 DOI: 10.1016/j.heliyon.2024.e38434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/15/2024] Open
Abstract
Quinoline-urea-benzothiazole hybrids exhibited low to sub-micromolar in vitro activities against the Plasmodium falciparum (P. falciparum) 3D7 chloroquine (CQ)-sensitive strain, with compounds 5a, 5b and 5f showing activities ranging from 0.33 to 0.97 μM. Against the formation of β-haematin, the majority of the tested compounds were comparable to the reference drug, chloroquine (CQ), with compounds 5c (IC50 = 9.55 ± 0.62 μM) and 5h (IC50 = 9.73 ± 1.38 μM), exhibiting slightly better in vitro efficacy than CQ. The hybrids also exhibited low micromolar to submicromolar activities against Trypanosoma brucei brucei, with 5j-5k being comparable to the reference drug, pentamidine. Compound 5b displayed higher in silico binding energy than CQ when docked against P. falciparum dihydroorotate dehydrogenase enzyme. Compounds 5j and 5k showed higher binding energies than pentamidine within the trypanothione reductase enzyme binding pocket. The root means square deviations of the hit compounds 5b, 5j and 5k were stable throughout the 100 ns simulation period. Post-molecular dynamics MMGBSA binding free energies showed that the selected hybrids bind spontaneously to the respective enzymes. The DFT investigation revealed that the compounds have regions that can bind to the electropositive and electronegative sites of the proteins.
Collapse
Affiliation(s)
- Oluwatoba E. Oyeneyin
- School of Chemistry and Physics, University of KwaZulu Natal, Westville Campus, Durban, 4000, South Africa
- Department of Chemical Sciences, Adekunle Ajasin University, Akungba-Akoko, Ondo State, Nigeria
| | - Rashmika Moodley
- School of Chemistry and Physics, University of KwaZulu Natal, Westville Campus, Durban, 4000, South Africa
| | - Chakes Mashaba
- School of Chemistry and Physics, University of KwaZulu Natal, Westville Campus, Durban, 4000, South Africa
| | - Larnelle F. Garnie
- Department of Chemistry, Faculty of Science, University of Cape Town, Rondebosch, Cape Town, 7700, South Africa
| | - Damilola A. Omoboyowa
- Department of Biochemistry, Adekunle Ajasin University, Akungba-Akoko, Ondo State, Nigeria
| | - Goitsemodimo H. Rakodi
- Department of Chemistry, Faculty of Science, Tshwane University of Technology, Pretoria, 001, South Africa
| | - Mabuatsela V. Maphoru
- Department of Chemistry, Faculty of Science, Tshwane University of Technology, Pretoria, 001, South Africa
| | - Mohamed O. Balogun
- Bio-Polymer Modification and Therapeutics Laboratory, Council for Scientific and Industrial Research (CSIR), Pretoria, 0001, South Africa
| | - Heinrich C. Hoppe
- Centre for Chemico- and Biomedical Research, Rhodes University, Makhanda, 6140, South Africa
- Department of Biochemistry and Microbiology, Faculty of Science, Rhodes University, Makhanda, 6140, South Africa
| | - Timothy J. Egan
- Department of Chemistry, Faculty of Science, University of Cape Town, Rondebosch, Cape Town, 7700, South Africa
- Institute of Infectious Diseases and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Rondebosch, 7701, South Africa
| | - Matshawandile Tukulula
- School of Chemistry and Physics, University of KwaZulu Natal, Westville Campus, Durban, 4000, South Africa
| |
Collapse
|
4
|
Oduselu GO, Elebiju OF, Ogunnupebi TA, Akash S, Ajani OO, Adebiyi E. Employing Hexahydroquinolines as PfCDPK4 Inhibitors to Combat Malaria Transmission: An Advanced Computational Approach. Adv Appl Bioinform Chem 2024; 17:83-105. [PMID: 39345873 PMCID: PMC11430315 DOI: 10.2147/aabc.s476404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 09/18/2024] [Indexed: 10/01/2024] Open
Abstract
Background Existing antimalarial drugs primarily target blood-stage parasites, but there is a need for transmission-blocking drugs to combat malaria effectively. Plasmodium falciparum Calcium-dependent Protein Kinase 4 (CDPK4) is a promising target for such drugs. This study employed advanced in silico analyses of hexahydroquinolines (HHQ) derivatives to identify PfCDPK4 inhibitors capable of disrupting malaria transmission. Structure-based virtual screening (SBVS) was employed to discover HHQ derivatives with the highest binding affinities against the 3D structure of PfCDPK4 (PDB 1D: 4QOX). Methods Interaction analysis of protein-ligand complexes utilized Discovery Studio Client, while druglikeness and ADMET properties were assessed using SwissADME and pkCSM web servers, respectively. Quantum mechanical calculations of the top hits were conducted using density functional theory (DFT), and GROMACS was employed to perform the molecular dynamics (MD) simulations. Binding free energy was predicted using the MMPBSA.py tool from the AMBER package. Results SBVS identified ten best hits possessing docking scores within the range of -11.2 kcal/mol and -10.6 kcal/mol, surpassing the known inhibitor, BKI-1294 (-9.9 kcal/mol). Among these, 4-[4-(Furan-2-carbonyl)piperazin-1-yl]-1-(naphthalen-2-ylmethyl)-2-oxo-4a,5,6,7,8,8a-hexahydroquinoline-3-carbonitrile (PubChem ID: 145784778) exhibited the highest binding affinity (-11.2 kcal/mol) against PfCDPK4. Conclusion Comparative analysis of this compound with BKI-1294 using advanced computational approaches demonstrated competitive potential. These findings suggest the potential of 4-[4-(Furan-2-carbonyl)piperazin-1-yl]-1-(naphthalen-2-ylmethyl)-2-oxo-4a,5,6,7,8,8a-hexahydroquinoline-3-carbonitrile as a promising PfCDPK4 inhibitor for disrupting malaria transmission. However, further experimental studies are warranted to validate its efficacy and safety profile.
Collapse
Affiliation(s)
- Gbolahan O Oduselu
- Covenant University Bioinformatics Research (CUBRe), Covenant University, Ota, OG, Nigeria
| | - Oluwadunni F Elebiju
- Covenant University Bioinformatics Research (CUBRe), Covenant University, Ota, OG, Nigeria
- Department of Chemistry, Covenant University, Ota, OG, Nigeria
| | - Temitope A Ogunnupebi
- Covenant University Bioinformatics Research (CUBRe), Covenant University, Ota, OG, Nigeria
- Department of Chemistry, Covenant University, Ota, OG, Nigeria
| | - Shopnil Akash
- Department of Pharmacy, Daffodil International University, Dhaka, Bangladesh
| | - Olayinka O Ajani
- Covenant University Bioinformatics Research (CUBRe), Covenant University, Ota, OG, Nigeria
- Department of Chemistry, Covenant University, Ota, OG, Nigeria
| | - Ezekiel Adebiyi
- Covenant University Bioinformatics Research (CUBRe), Covenant University, Ota, OG, Nigeria
- African Center of Excellence in Bioinformatics & Data Intensive Science, Makerere University, Kampala, Uganda
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
5
|
Van Horn KS, Zhao Y, Parvatkar PT, Maier J, Mutka T, Lacrue A, Brockmeier F, Ebert D, Wu W, Casandra DR, Namelikonda N, Yacoub J, Sigal M, Knapp S, Floyd D, Waterson D, Burrows JN, Duffy J, DeRisi JL, Kyle DE, Guy RK, Manetsch R. Optimization of diastereomeric dihydropyridines as antimalarials. Eur J Med Chem 2024; 275:116599. [PMID: 38909569 DOI: 10.1016/j.ejmech.2024.116599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 06/04/2024] [Accepted: 06/12/2024] [Indexed: 06/25/2024]
Abstract
The increase in research funding for the development of antimalarials since 2000 has led to a surge of new chemotypes with potent antimalarial activity. High-throughput screens have delivered several thousand new active compounds in several hundred series, including the 4,7-diphenyl-1,4,5,6,7,8-hexahydroquinolines, hereafter termed dihydropyridines (DHPs). We optimized the DHPs for antimalarial activity. Structure-activity relationship studies focusing on the 2-, 3-, 4-, 6-, and 7-positions of the DHP core led to the identification of compounds potent (EC50 < 10 nM) against all strains of P. falciparum tested, including the drug-resistant parasite strains K1, W2, and TM90-C2B. Evaluation of efficacy of several compounds in vivo identified two compounds that reduced parasitemia by >75 % in mice 6 days post-exposure following a single 50 mg/kg oral dose. Resistance acquisition experiments with a selected dihydropyridine led to the identification of a single mutation conveying resistance in the gene encoding for Plasmodium falciparum multi-drug resistance protein 1 (PfMDR1). The same dihydropyridine possessed transmission blocking activity. The DHPs have the potential for the development of novel antimalarial drug candidates.
Collapse
Affiliation(s)
- Kurt S Van Horn
- Department of Chemistry, University of South Florida, 4202 E. Fowler Avenue, Tampa, FL, 33620, United States; Department of Chemistry and Chemical Biology, Northeastern University, 360 Huntington Avenue, Boston, MA, 02115, United States.
| | - Yingzhao Zhao
- Department of Chemistry and Chemical Biology, Northeastern University, 360 Huntington Avenue, Boston, MA, 02115, United States
| | - Prakash T Parvatkar
- Department of Chemistry and Chemical Biology, Northeastern University, 360 Huntington Avenue, Boston, MA, 02115, United States
| | - Julie Maier
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, United States
| | - Tina Mutka
- Department of Global Health, College of Public Health, University of South Florida, 3720 Spectrum Boulevard, Tampa, FL, 33612, United States
| | - Alexis Lacrue
- Department of Global Health, College of Public Health, University of South Florida, 3720 Spectrum Boulevard, Tampa, FL, 33612, United States
| | - Fabian Brockmeier
- Department of Chemistry and Chemical Biology, Northeastern University, 360 Huntington Avenue, Boston, MA, 02115, United States
| | - Daniel Ebert
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, 94158, United States
| | - Wesley Wu
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, 94158, United States
| | - Debora R Casandra
- Department of Global Health, College of Public Health, University of South Florida, 3720 Spectrum Boulevard, Tampa, FL, 33612, United States
| | - Niranjan Namelikonda
- Department of Chemistry, University of South Florida, 4202 E. Fowler Avenue, Tampa, FL, 33620, United States
| | - Jeanine Yacoub
- Department of Chemistry, University of South Florida, 4202 E. Fowler Avenue, Tampa, FL, 33620, United States
| | - Martina Sigal
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, United States
| | - Spencer Knapp
- Department of Chemistry and Chemical Biology, Rutgers - The State University of New Jersey, 610 Taylor Road, Piscataway, NJ, 08854, United States
| | - David Floyd
- Department of Chemistry and Chemical Biology, Rutgers - The State University of New Jersey, 610 Taylor Road, Piscataway, NJ, 08854, United States
| | - David Waterson
- Medicines for Malaria Venture, 20, Route de Pré-Bois, P.O. Box 1826, 1215, Geneva, 15, Switzerland
| | - Jeremy N Burrows
- Medicines for Malaria Venture, 20, Route de Pré-Bois, P.O. Box 1826, 1215, Geneva, 15, Switzerland
| | - James Duffy
- Medicines for Malaria Venture, 20, Route de Pré-Bois, P.O. Box 1826, 1215, Geneva, 15, Switzerland
| | - Joseph L DeRisi
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, 94158, United States; Howard Hughes Medical Institute, Chevy Chase, MD, 20815, United States
| | - Dennis E Kyle
- Department of Global Health, College of Public Health, University of South Florida, 3720 Spectrum Boulevard, Tampa, FL, 33612, United States; Center for Tropical & Emerging Global Diseases, University of Georgia, Athens, GA, 30602, United States
| | - R Kiplin Guy
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, United States; Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, 40506, United States
| | - Roman Manetsch
- Department of Chemistry, University of South Florida, 4202 E. Fowler Avenue, Tampa, FL, 33620, United States; Department of Chemistry and Chemical Biology, Northeastern University, 360 Huntington Avenue, Boston, MA, 02115, United States; Department of Pharmaceutical Sciences, Northeastern University, 360 Huntington Avenue, Boston, MA, 02115, United States.
| |
Collapse
|
6
|
Amorzesh H, Bayat M, Nasri S. Catalyst-free synthesis of highly functionalized triazole hexahydroquinoline carbohydrazide scaffolds via four-component cyclocondensation reaction. Mol Divers 2024; 28:51-60. [PMID: 36585569 DOI: 10.1007/s11030-022-10592-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 12/22/2022] [Indexed: 01/01/2023]
Abstract
A new class of multi-functional triazole hexahydroquinoline carbohydrazide named 2-amino-7,7-dimethyl-5-oxo-4-phenyl-1-(4H-1,2,4-triazol-3-yl)-1,4,5,6,7,8-hexahydroquinoline-3-carbohydrazide has been synthesized by a novel multi-component process involving the reaction of dimedone, 3-amino-1,2,4-triazole, various benzaldehyde with cyanoacetohydrazide under mild conditions in the stoichiometric melt and chloroform in sequence. The simple one-pot process, straight product isolation without applying tedious purification procedures, progression of the reaction without using any catalyst, the application of diverse aldehydes causing a high molecular diversity, the existence of several nitrogen atoms in the product structure, and the possibility of creating multiple hydrogen bonding in the final compound are attractive specifications of the present strategy.
Collapse
Affiliation(s)
- Hasti Amorzesh
- Department of Chemistry, Faculty of Science, Imam Khomeini International University, Qazvin, Iran
| | - Mohammad Bayat
- Department of Chemistry, Faculty of Science, Imam Khomeini International University, Qazvin, Iran.
| | - Shima Nasri
- Department of Chemistry, Faculty of Science, Imam Khomeini International University, Qazvin, Iran
| |
Collapse
|
7
|
Erhunse N, Kumari S, Anmol, Singh P, Omoregie ES, Singh AP, Sharma U, Sahal D. Annickia affinis (Exell) Versteegh & Sosef methanol stem bark extract, potent fractions and isolated Berberine alkaloid target both blood and liver stages of malaria parasites. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117269. [PMID: 37813288 DOI: 10.1016/j.jep.2023.117269] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 09/28/2023] [Accepted: 09/30/2023] [Indexed: 10/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Having identified Annickia affinis as the most potent antiplasmodial plant constituent in a hepta-herbal Agbo-iba (HHA) formula commonly used to manage malaria in Benin city, Nigeria, we have in this study attempted to identify the specialized metabolites responsible for antiplasmodial activity of A. affinis through anti-blood stage malaria parasite activity guided isolation of potent molecules from its stem bark methanol extract. After that, phenotypic effects, including stage-specific kill kinetics, were investigated. Further, the crude extract, its potent fractions, and specialized metabolites were also tested against the liver-stage malaria parasite. MATERIALS AND METHODS A. affinis was subjected to molecular PCR-based analysis to confirm its identity. Thereafter, extraction of its stem bark with methanol was carried out. Alkaloid enriched fractions from this stem bark extract were obtained using the acid-base-solvent extraction method. These alkaloid-enriched fractions were subjected to various chromatographic techniques that led to the isolation of two protoberberine alkaloids identified as berberine and palmatine based on NMR and mass spectrometry analysis. The efficacy of crude extract, fractions and purified alkaloids was tested against the malaria parasite's blood and liver stages, respectively. RESULTS AND DISCUSSION Annickia affinis methanol extract, fractions, and the isolated protoberberine alkaloids showed excellent antiplasmodial activity with good selectivity against blood-stage malaria parasite. Thus, their IC50 against various strains of the parasite ranged from 0.95 to 18.65 μg/ml, while CC50 against Human embryonic kidney (HEK) and the human hepatoma (HUH-7) cell lines ranged between 10 and > 100 μg/ml. Interestingly, the crude extract and the alkaloid enriched fractions showed promising activity against the liver-stage malaria parasite. Between berberine and palmatine isolated from the potent fractions, only the former showed ∼100% and 90% inhibitions of liver stage parasite at 5 μg/ml and 1 μg/ml, respectively, while the latter showed no inhibition even at 20 μg/ml. CONCLUSION This study reports that the ethnomedicinal use of HHA to manage malaria can be attributed to the presence of promising antiplasmodial protoberberine alkaloids together with synergistic effects via either enhancement of bioavailability or improved pharmacokinetics by other phytoconstituent(s) coming from other HHA constituent plants. The protoberberine alkaloids isolated have been identified as fast-acting antiplasmodial agents, with activity against all erythrocytic stages of the malaria parasite. Further, A. affinis methanol stembark extract and the protoberberine alkaloid berberine isolated from it also displayed excellent activity (>90% inhibition at 1 μg/ml) against the liver-stage malaria parasite. A. affinis and HHA can thus be useful as both liver-stage prophylactic and blood-stage curative agents.
Collapse
Affiliation(s)
- Nekpen Erhunse
- Malaria Drug Discovery Research Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, 110067, India; Department of Biochemistry, Faculty of Life Sciences, University of Benin, Benin city, Nigeria
| | - Surekha Kumari
- Chemical Technology Division CSIR-IHBT, Palampur, Himachal Pradesh, 176061, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Anmol
- Chemical Technology Division CSIR-IHBT, Palampur, Himachal Pradesh, 176061, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Pooja Singh
- Infectious Diseases Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhiz, 110067, India
| | | | - Agam Prasad Singh
- Infectious Diseases Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhiz, 110067, India
| | - Upendra Sharma
- Chemical Technology Division CSIR-IHBT, Palampur, Himachal Pradesh, 176061, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Dinkar Sahal
- Malaria Drug Discovery Research Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, 110067, India.
| |
Collapse
|
8
|
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
|
9
|
Muema JM, Mutunga JM, Obonyo MA, Getahun MN, Mwakubambanya RS, Akala HM, Cheruiyot AC, Yeda RA, Juma DW, Andagalu B, Johnson JL, Roth AL, Bargul JL. Isoliensinine from Cissampelos pariera rhizomes exhibits potential gametocytocidal and anti-malarial activities against Plasmodium falciparum clinical isolates. Malar J 2023; 22:161. [PMID: 37208735 DOI: 10.1186/s12936-023-04590-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 05/15/2023] [Indexed: 05/21/2023] Open
Abstract
BACKGROUND The unmet demand for effective malaria transmission-blocking agents targeting the transmissible stages of Plasmodium necessitates intensive discovery efforts. In this study, a bioactive bisbenzylisoquinoline (BBIQ), isoliensinine, from Cissampelos pariera (Menispermaceae) rhizomes was identified and characterized for its anti-malarial activity. METHODS Malaria SYBR Green I fluorescence assay was performed to evaluate the in vitro antimalarial activity against D6, Dd2, and F32-ART5 clones, and immediate ex vivo (IEV) susceptibility for 10 freshly collected P. falciparum isolates. To determine the speed- and stage-of-action of isoliensinine, an IC50 speed assay and morphological analyses were performed using synchronized Dd2 asexuals. Gametocytocidal activity against two culture-adapted gametocyte-producing clinical isolates was determined using microscopy readouts, with possible molecular targets and their binding affinities deduced in silico. RESULTS Isoliensinine displayed a potent in vitro gametocytocidal activity at mean IC50gam values ranging between 0.41 and 0.69 µM for Plasmodium falciparum clinical isolates. The BBIQ compound also inhibited asexual replication at mean IC50Asexual of 2.17 µM, 2.22 µM, and 2.39 µM for D6, Dd2 and F32-ART5 respectively, targeting the late-trophozoite to schizont transition. Further characterization demonstrated a considerable immediate ex vivo potency against human clinical isolates at a geometric mean IC50IEV = 1.433 µM (95% CI 0.917-2.242). In silico analyses postulated a probable anti-malarial mechanism of action by high binding affinities for four mitotic division protein kinases; Pfnek1, Pfmap2, Pfclk1, and Pfclk4. Additionally, isoliensinine was predicted to possess an optimal pharmacokinetics profile and drug-likeness properties. CONCLUSION These findings highlight considerable grounds for further exploration of isoliensinine as an amenable scaffold for malaria transmission-blocking chemistry and target validation.
Collapse
Affiliation(s)
- Jackson M Muema
- Department of Biochemistry, Jomo Kenyatta University of Agriculture and Technology (JKUAT), Nairobi, Kenya.
- U.S. Army Medical Research Directorate-Africa (USAMRD-A), Centre for Global Health Research (CGHR), Kenya Medical Research Institute (KEMRI), Kisumu, Kenya.
| | - James M Mutunga
- U.S. Army Medical Research Directorate-Africa (USAMRD-A), Centre for Global Health Research (CGHR), Kenya Medical Research Institute (KEMRI), Kisumu, Kenya
- Department of Biological Sciences, School of Pure and Applied Sciences, Mount Kenya University, Thika, Kenya
- School of Engineering Design, Technology and Professional Programs, Pennsylvania State University, University Park, PA, 16802, USA
| | - Meshack A Obonyo
- Department of Biochemistry and Molecular Biology, Egerton University, Egerton, Kenya
| | - Merid N Getahun
- International Centre of Insect Physiology and Ecology (Icipe), Nairobi, Kenya
| | | | - Hoseah M Akala
- U.S. Army Medical Research Directorate-Africa (USAMRD-A), Centre for Global Health Research (CGHR), Kenya Medical Research Institute (KEMRI), Kisumu, Kenya
| | - Agnes C Cheruiyot
- U.S. Army Medical Research Directorate-Africa (USAMRD-A), Centre for Global Health Research (CGHR), Kenya Medical Research Institute (KEMRI), Kisumu, Kenya
| | - Redemptah A Yeda
- U.S. Army Medical Research Directorate-Africa (USAMRD-A), Centre for Global Health Research (CGHR), Kenya Medical Research Institute (KEMRI), Kisumu, Kenya
| | - Dennis W Juma
- U.S. Army Medical Research Directorate-Africa (USAMRD-A), Centre for Global Health Research (CGHR), Kenya Medical Research Institute (KEMRI), Kisumu, Kenya
| | - Ben Andagalu
- U.S. Army Medical Research Directorate-Africa (USAMRD-A), Centre for Global Health Research (CGHR), Kenya Medical Research Institute (KEMRI), Kisumu, Kenya
| | - Jaree L Johnson
- U.S. Army Medical Research Directorate-Africa (USAMRD-A), Centre for Global Health Research (CGHR), Kenya Medical Research Institute (KEMRI), Kisumu, Kenya
| | - Amanda L Roth
- U.S. Army Medical Research Directorate-Africa (USAMRD-A), Centre for Global Health Research (CGHR), Kenya Medical Research Institute (KEMRI), Kisumu, Kenya
| | - Joel L Bargul
- Department of Biochemistry, Jomo Kenyatta University of Agriculture and Technology (JKUAT), Nairobi, Kenya.
- International Centre of Insect Physiology and Ecology (Icipe), Nairobi, Kenya.
| |
Collapse
|
10
|
Nourani L, Mehrizi AA, Pirahmadi S, Pourhashem Z, Asadollahi E, Jahangiri B. CRISPR/Cas advancements for genome editing, diagnosis, therapeutics, and vaccine development for Plasmodium parasites, and genetic engineering of Anopheles mosquito vector. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2023; 109:105419. [PMID: 36842543 DOI: 10.1016/j.meegid.2023.105419] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/30/2023] [Accepted: 02/21/2023] [Indexed: 02/28/2023]
Abstract
Malaria as vector-borne disease remains important health concern with over 200 million cases globally. Novel antimalarial medicines and more effective vaccines must be developed to eliminate and eradicate malaria. Appraisal of preceding genome editing approaches confirmed the CRISPR/Cas nuclease system as a novel proficient genome editing system and a tool for species-specific diagnosis, and drug resistance researches for Plasmodium species, and gene drive to control Anopheles population. CRISPR/Cas technology, as a handy tool for genome editing can be justified for the production of transgenic malaria parasites like Plasmodium transgenic lines expressing Cas9, chimeric Plasmodium transgenic lines, knockdown and knockout transgenic parasites, and transgenic parasites expressing alternative alleles, and also mutant strains of Anopheles such as only male mosquito populations, generation of wingless mosquitoes, and creation of knock-out/ knock-in mutants. Though, the incorporation of traditional methods and novel molecular techniques could noticeably enhance the quality of results. The striking development of a CRISPR/Cas-based diagnostic kit that can specifically diagnose the Plasmodium species or drug resistance markers is highly required in malaria settings with affordable cost and high-speed detection. Furthermore, the advancement of genome modifications by CRISPR/Cas technologies resolves contemporary restrictions to culturing, maintaining, and analyzing these parasites, and the aptitude to investigate parasite genome functions opens up new vistas in the better understanding of pathogenesis.
Collapse
Affiliation(s)
- Leila Nourani
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Akram Abouie Mehrizi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran.
| | - Sakineh Pirahmadi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Zeinab Pourhashem
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Elahe Asadollahi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| | - Babak Jahangiri
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
11
|
Liu L, Zhang Z, Liu H, Zhu S, Zhou T, Wang C, Hu M. Identification and characterisation of the haemozoin of Haemonchus contortus. Parasit Vectors 2023; 16:88. [PMID: 36879311 PMCID: PMC9990328 DOI: 10.1186/s13071-023-05714-3] [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: 01/20/2023] [Accepted: 02/22/2023] [Indexed: 03/08/2023] Open
Abstract
BACKGROUND Most haematophagous organisms constantly suck the host's haemoglobin, which produces toxic free haem. This toxic haem aggregation into the nontoxic crystallisation complex known as haemozoin represents one of the most important detoxification pathways in living organisms, but very little is known about the features of haemozoin in parasitic nematodes. Here, we identified and characterised the haemozoin of an economically significant blood-sucking nematode, Haemonchus contortus. METHODS Using electron microscopy, spectrophotometry analyses and biochemical approaches, haemozoin crystallisation was identified and characterised in parasitic fourth-stage larvae (L4s) and/or adult worms as well as L4s of in vitro culture. RESULTS The haemozoin was formed in intestinal lipid droplets of the parasitic L4s and adult worms. The characterisation of the haemozoin showed regularly spherical structures and had a 400-nm absorption peak. Furthermore, the haemozoin in in vitro cultured L4s was associated with the culture time and concentration of red blood cells added into the medium, and its formation could be inhibited by chloroquine-derived drugs. CONCLUSIONS This work provides detailed insight into the haemozoin formation of H. contortus and should have important implications for developing novel therapeutic targets against this parasite or related haematophagous organisms.
Collapse
Affiliation(s)
- Lu Liu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Zongshan Zhang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Hui Liu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Shengnan Zhu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Taoxun Zhou
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China
| | - Chunqun Wang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Min Hu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
12
|
Camargo da Luz L, Gözde Gündüz M, Beal R, Modernell Zanotto G, Ramires Kuhn E, Augusto Netz P, Şafak C, Fernando Bruno Gonçalves P, da Silveira Santos F, Severo Rodembusch F. Theoretical and experimental investigation of 1,4-dihydropyridine-based hexahydroquinoline-3-carboxylates: Photophysics and bovine serum albumin binding studies. J Photochem Photobiol A Chem 2022. [DOI: 10.1016/j.jphotochem.2022.113915] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
13
|
Ramjith J, Alkema M, Bradley J, Dicko A, Drakeley C, Stone W, Bousema T. Quantifying Reductions in Plasmodium falciparum Infectivity to Mosquitos: A Sample Size Calculator to Inform Clinical Trials on Transmission-Reducing Interventions. Front Immunol 2022; 13:899615. [PMID: 35720362 PMCID: PMC9205189 DOI: 10.3389/fimmu.2022.899615] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 05/10/2022] [Indexed: 11/13/2022] Open
Abstract
Malaria transmission depends on the presence of mature Plasmodium transmission stages (gametocytes) that may render blood-feeding Anopheles mosquitos infectious. Transmission-blocking antimalarial drugs and vaccines can prevent transmission by reducing gametocyte densities or infectivity to mosquitos. Mosquito infection outcomes are thereby informative biological endpoints of clinical trials with transmission blocking interventions. Nevertheless, trials are often primarily designed to determine intervention safety; transmission blocking efficacy is difficult to incorporate in sample size considerations due to variation in infection outcomes and considerable inter-study variation. Here, we use clinical trial data from studies in malaria naive and naturally exposed study participants to present an online sample size calculator tool. This sample size calculator allows studies to be powered to detect reductions in the proportion of infected mosquitos or infection burden (oocyst density) in mosquitos. The utility of this online tool is illustrated using trial data with transmission blocking malaria drugs.
Collapse
Affiliation(s)
- Jordache Ramjith
- Radboud Institute for Molecular Life Sciences, Department of Medical Microbiology, Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
- Department for Health Evidence, Biostatistics Research Group, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Manon Alkema
- Radboud Institute for Molecular Life Sciences, Department of Medical Microbiology, Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - John Bradley
- Medical Research Council (MRC) International Statistics and Epidemiology Group, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Alassane Dicko
- Malaria Research and Training Centre, Faculty of Pharmacy and Faculty of Medicine and Dentistry, University of Science, Techniques and Technologies of Bamako, Bamako, Mali
| | - Chris Drakeley
- Medical Research Council (MRC) International Statistics and Epidemiology Group, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Will Stone
- Medical Research Council (MRC) International Statistics and Epidemiology Group, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Teun Bousema
- Radboud Institute for Molecular Life Sciences, Department of Medical Microbiology, Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
14
|
Cheng P, Zhang Z, Yang F, Cai S, Wang L, Wang C, Wang M, Liu Y, Fei C, Zhang L, Xue F, Gu F. FnCas12a/crRNA-Mediated Genome Editing in Eimeria tenella. Front Genet 2021; 12:738746. [PMID: 34630528 PMCID: PMC8494306 DOI: 10.3389/fgene.2021.738746] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 08/31/2021] [Indexed: 12/16/2022] Open
Abstract
Eimeria species are intracellular parasites residing inside the intestinal epithelial cell, which cause poultry coccidiosis and result in significant financial losses in the poultry industry. Genome editing of Eimeria is of immense importance for the development of vaccines and drugs. CRISPR/Cas9 has been utilized for manipulating the genome of Eimeria tenella (E. tenella). Ectopic expression of Cas9, i.e., via plasmids, would introduce transgene, which substantially limits its application, especially for vaccine development. In this study, we initially optimized the condition of the transfection protocol. We demonstrated that with the optimized condition, the transfection of FnCas12a (also known as "FnCpf1") protein and crRNA targeting EtHistone H4 triggered DNA double-strand breaks in vivo. We then used this strategy to knock-in a coding cassette for an enhanced yellow fluorescent protein (EYFP) and dihydrofolate reductase-thymidylate synthase gene (DHFR) as a selection marker to tag endogenous EtActin. The engineered E. tenella parasite possesses EYFP expression in its entire life cycle. Our results demonstrated that FnCas12a could trigger genome editing in E. tenella, which augments the applicability of the dissection of gene function and the development of anticoccidial drugs and vaccines for Eimeria species.
Collapse
Affiliation(s)
- Peipei Cheng
- Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Zhihao Zhang
- Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Fayu Yang
- Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Shuo Cai
- Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Lina Wang
- Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Chunmei Wang
- Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Mi Wang
- Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Yingchun Liu
- Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Chenzhong Fei
- Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Lifang Zhang
- Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Feiqun Xue
- Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Feng Gu
- Key Laboratory of Veterinary Chemical Drugs and Pharmaceutics, Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| |
Collapse
|
15
|
Erhunse N, Sahal D. Protecting future antimalarials from the trap of resistance: Lessons from artemisinin-based combination therapy (ACT) failures. J Pharm Anal 2021; 11:541-554. [PMID: 34765267 PMCID: PMC8572664 DOI: 10.1016/j.jpha.2020.07.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 07/19/2020] [Accepted: 07/19/2020] [Indexed: 11/01/2022] Open
Abstract
Having faced increased clinical treatment failures with dihydroartemisinin-piperaquine (DHA-PPQ), Cambodia swapped the first line artemisinin-based combination therapy (ACT) from DHA-PPQ to artesunate-mefloquine given that parasites resistant to piperaquine are susceptible to mefloquine. However, triple mutants have now emerged, suggesting that drug rotations may not be adequate to keep resistance at bay. There is, therefore, an urgent need for alternative treatment strategies to tackle resistance and prevent its spread. A proper understanding of all contributors to artemisinin resistance may help us identify novel strategies to keep artemisinins effective until new drugs become available for their replacement. This review highlights the role of the key players in artemisinin resistance, the current strategies to deal with it and suggests ways of protecting future antimalarial drugs from bowing to resistance as their predecessors did.
Collapse
Affiliation(s)
- Nekpen Erhunse
- Malaria Drug Discovery Research Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, 110067, India
- Department of Biochemistry, Faculty of Life Sciences, University of Benin, Benin City, Edo-State, Nigeria
| | - Dinkar Sahal
- Malaria Drug Discovery Research Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, 110067, India
| |
Collapse
|
16
|
Murithi JM, Pascal C, Bath J, Boulenc X, Gnädig NF, Pasaje CFA, Rubiano K, Yeo T, Mok S, Klieber S, Desert P, Jiménez-Díaz MB, Marfurt J, Rouillier M, Cherkaoui-Rbati MH, Gobeau N, Wittlin S, Uhlemann AC, Price RN, Wirjanata G, Noviyanti R, Tumwebaze P, Cooper RA, Rosenthal PJ, Sanz LM, Gamo FJ, Joseph J, Singh S, Bashyam S, Augereau JM, Giraud E, Bozec T, Vermat T, Tuffal G, Guillon JM, Menegotto J, Sallé L, Louit G, Cabanis MJ, Nicolas MF, Doubovetzky M, Merino R, Bessila N, Angulo-Barturen I, Baud D, Bebrevska L, Escudié F, Niles JC, Blasco B, Campbell S, Courtemanche G, Fraisse L, Pellet A, Fidock DA, Leroy D. The antimalarial MMV688533 provides potential for single-dose cures with a high barrier to Plasmodium falciparum parasite resistance. Sci Transl Med 2021; 13:13/603/eabg6013. [PMID: 34290058 PMCID: PMC8530196 DOI: 10.1126/scitranslmed.abg6013] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 07/02/2021] [Indexed: 01/13/2023]
Abstract
The emergence and spread of Plasmodium falciparum resistance to first-line antimalarials creates an imperative to identify and develop potent preclinical candidates with distinct modes of action. Here, we report the identification of MMV688533, an acylguanidine that was developed following a whole-cell screen with compounds known to hit high-value targets in human cells. MMV688533 displays fast parasite clearance in vitro and is not cross-resistant with known antimalarials. In a P. falciparum NSG mouse model, MMV688533 displays a long-lasting pharmacokinetic profile and excellent safety. Selection studies reveal a low propensity for resistance, with modest loss of potency mediated by point mutations in PfACG1 and PfEHD. These proteins are implicated in intracellular trafficking, lipid utilization, and endocytosis, suggesting interference with these pathways as a potential mode of action. This preclinical candidate may offer the potential for a single low-dose cure for malaria.
Collapse
Affiliation(s)
- James M. Murithi
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Cécile Pascal
- Sanofi, Infectious Diseases Therapeutic Area, Marcy l'Etoile, France
| | - Jade Bath
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | | | - Nina F. Gnädig
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | | | - Kelly Rubiano
- Department of Microbiology and Immunology, 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
| | - Sachel Mok
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Sylvie Klieber
- Sanofi R&D, Translational Medicine & Early Development, Montpellier, France
| | | | | | - Jutta Marfurt
- Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University, Darwin, Australia
| | | | | | | | - Sergio Wittlin
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Basel, Switzerland.,Universität Basel, Basel, Switzerland
| | - Anne-Catrin Uhlemann
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Ric N. Price
- Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University, Darwin, Australia.,Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK.,Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Grennady Wirjanata
- Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University, Darwin, Australia
| | | | | | - Roland A. Cooper
- Department of Natural Sciences and Mathematics, Dominican University of California, San Rafael, CA, USA
| | | | - Laura M. Sanz
- Global Health Pharma Research Unit, GSK, Tres Cantos, Madrid, Spain
| | | | | | | | | | | | - Elie Giraud
- Sanofi, Infectious Diseases Therapeutic Area, Marcy l'Etoile, France
| | - Tanguy Bozec
- Sanofi, Infectious Diseases Therapeutic Area, Marcy l'Etoile, France
| | - Thierry Vermat
- Sanofi, Infectious Diseases Therapeutic Area, Marcy l'Etoile, France
| | - Gilles Tuffal
- Sanofi R&D, Translational Medicine & Early Development, Montpellier, France
| | | | - Jérôme Menegotto
- Sanofi, Infectious Diseases Therapeutic Area, Marcy l'Etoile, France
| | - Laurent Sallé
- Sanofi R&D, Translational Medicine & Early Development, Montpellier, France
| | | | - Marie-José Cabanis
- Sanofi R&D, Translational Medicine & Early Development, Montpellier, France
| | | | | | - Rita Merino
- Sanofi, Infectious Diseases Therapeutic Area, Marcy l'Etoile, France
| | - Nadir Bessila
- Sanofi, Infectious Diseases Therapeutic Area, Marcy l'Etoile, France
| | | | | | | | | | - Jacquin C. Niles
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | | | | | - Laurent Fraisse
- Sanofi, Infectious Diseases Therapeutic Area, Marcy l'Etoile, France
| | - Alain Pellet
- Sanofi, Infectious Diseases Therapeutic Area, Marcy l'Etoile, France
| | - David A. Fidock
- Department of Microbiology and Immunology, 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.,Corresponding author. (D.A.F.); (D.L.)
| | - Didier Leroy
- Medicines for Malaria Venture, Geneva, Switzerland.,Corresponding author. (D.A.F.); (D.L.)
| |
Collapse
|
17
|
de Villiers KA, Egan TJ. Heme Detoxification in the Malaria Parasite: A Target for Antimalarial Drug Development. Acc Chem Res 2021; 54:2649-2659. [PMID: 33982570 DOI: 10.1021/acs.accounts.1c00154] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Over the last century, malaria deaths have decreased by more than 85%. Nonetheless, there were 405 000 deaths in 2018, mostly resulting from Plasmodium falciparum infection. In the 21st century, much of the advance has arisen from the deployment of insecticide-treated bed nets and artemisinin combination therapy. However, over the past few decades parasites with a delayed artemisinin clearance phenotype have appeared in Southeast Asia, threatening further gains. The effort to find new drugs is thus urgent. A prominent process in blood stage malaria parasites, which we contend remains a viable drug target, is hemozoin formation. This crystalline material consisting of heme can be readily seen when parasites are viewed microscopically. The process of its formation in the parasite, however, is still not fully understood.In early work, we recognized hemozoin formation as a biomineralization process. We have subsequently investigated the kinetics of synthetic hemozoin (β-hematin) crystallization catalyzed at lipid-aqueous interfaces under biomimetic conditions. This led us to the use of neutral detergent-based high-throughput screening (HTS) for inhibitors of β-hematin formation. A good hit rate against malaria parasites was obtained. Simultaneously, we developed a pyridine-based assay which proved successful in measuring the concentrations of hematin not converted to β-hematin.The pyridine assay was adapted to determine the effects of chloroquine and other clinical antimalarials on hemozoin formation in the cell. This permitted the determination of the dose-dependent amounts of exchangeable heme and hemozoin in P. falciparum for the first time. These studies have shown that hemozoin inhibitors cause a dose-dependent increase in exchangeable heme, correlated with decreased parasite survival. Electron spectroscopic imaging (ESI) showed a relocation of heme iron into the parasite cytoplasm, while electron microscopy provided evidence of the disruption of hemozoin crystals. This cellular assay was subsequently extended to top-ranked hits from a wide range of scaffolds found by HTS. Intriguingly, the amounts of exchangeable heme at the parasite growth IC50 values of these scaffolds showed substantial variation. The amount of exchangeable heme was found to be correlated with the amount of inhibitor accumulated in the parasitized red blood cell. This suggests that heme-inhibitor complexes, rather than free heme, lead to parasite death. This was supported by ESI using a Br-containing compound which showed the colocalization of Fe and Br as well as by confocal Raman microscopy which confirmed the presence of a complex in the parasite. Current evidence indicates that inhibitors block hemozoin formation by surface adsorption. Indeed, we have successfully introduced molecular docking with hemozoin to find new inhibitors. It follows that the resulting increase in free heme leads to the formation of the parasiticidal heme-inhibitor complex. We have reported crystal structures of heme-drug complexes for several aryl methanol antimalarials in nonaqueous media. These form coordination complexes but most other inhibitors interact noncovalently, and the determination of their structures remains a major challenge.It is our view that key future developments will include improved assays to measure cellular heme levels, better in silico approaches for predicting β-hematin inhibition, and a concerted effort to determine the structure and properties of heme-inhibitor complexes.
Collapse
Affiliation(s)
- Katherine A. de Villiers
- Department of Chemistry and Polymer Science, Stellenbosch University, Private Bag, Matieland 7600, South Africa
| | - Timothy J. Egan
- Department of Chemistry, University of Cape Town, Private Bag, Rondebosch 7701, South Africa
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, Cape Town 7945, South Africa
| |
Collapse
|
18
|
Okombo J, Kanai M, Deni I, Fidock DA. Genomic and Genetic Approaches to Studying Antimalarial Drug Resistance and Plasmodium Biology. Trends Parasitol 2021; 37:476-492. [PMID: 33715941 PMCID: PMC8162148 DOI: 10.1016/j.pt.2021.02.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 02/11/2021] [Accepted: 02/12/2021] [Indexed: 12/14/2022]
Abstract
Recent progress in genomics and molecular genetics has empowered novel approaches to study gene functions in disease-causing pathogens. In the human malaria parasite Plasmodium falciparum, the application of genome-based analyses, site-directed genome editing, and genetic systems that allow for temporal and quantitative regulation of gene and protein expression have been invaluable in defining the genetic basis of antimalarial resistance and elucidating candidate targets to accelerate drug discovery efforts. Using examples from recent studies, we review applications of some of these approaches in advancing our understanding of Plasmodium biology and illustrate their contributions and limitations in characterizing parasite genomic loci associated with antimalarial drug responses.
Collapse
Affiliation(s)
- John Okombo
- Department of Microbiology & Immunology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Mariko Kanai
- Department of Microbiology & Immunology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Ioanna Deni
- Department of Microbiology & Immunology, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - David A Fidock
- Department of Microbiology & Immunology, Department of Medicine, 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
|
19
|
Duffey M, Blasco B, Burrows JN, Wells TNC, Fidock DA, Leroy D. Assessing risks of Plasmodium falciparum resistance to select next-generation antimalarials. Trends Parasitol 2021; 37:709-721. [PMID: 34001441 DOI: 10.1016/j.pt.2021.04.006] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 04/01/2021] [Accepted: 04/08/2021] [Indexed: 02/07/2023]
Abstract
Strategies to counteract or prevent emerging drug resistance are crucial for the design of next-generation antimalarials. In the past, resistant parasites were generally identified following treatment failures in patients, and compounds would have to be abandoned late in development. An early understanding of how candidate therapeutics lose efficacy as parasites evolve resistance is important to facilitate drug design and improve resistance detection and monitoring up to the postregistration phase. We describe a new strategy to assess resistance to antimalarial compounds as early as possible in preclinical development by leveraging tools to define the Plasmodium falciparum resistome, predict potential resistance risks of clinical failure for candidate therapeutics, and inform decisions to guide antimalarial drug development.
Collapse
Affiliation(s)
| | - Benjamin Blasco
- Medicines for Malaria Venture, Geneva, Switzerland; Global Antibiotic Research and Development Partnership, Geneva, Switzerland
| | | | | | - David A Fidock
- Department of Microbiology and Immunology, 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
| | - Didier Leroy
- Medicines for Malaria Venture, Geneva, Switzerland.
| |
Collapse
|
20
|
Kirti A, Sharma M, Rani K, Bansal A. CRISPRing protozoan parasites to better understand the biology of diseases. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2021; 180:21-68. [PMID: 33934837 DOI: 10.1016/bs.pmbts.2021.01.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Precise gene editing techniques are paramount to gain deeper insights into the biological processes such as host-parasite interactions, drug resistance mechanisms, and gene-function relationships. Discovery of CRISPR-Cas9 system has spearheaded mechanistic understanding of protozoan parasite biology as evident from the number of reports in the last decade. Here, we have described the use of CRISPR-Cas9 in understanding the biology of medically important protozoan parasites such as Plasmodium, Leishmania, Trypanosoma, Babesia and Trichomonas. In spite of intrinsic difficulties in genome editing in these protozoan parasites, CRISPR-Cas9 has acted as a catalyst for faster generation of desired transgenic parasites. Modifications in the CRISPR-Cas9 system for improving the efficiency have been useful in better understanding the molecular mechanisms associated with repair of double strand breaks in the parasites. Moreover, improvement in reagents used for CRISPR mediated gene editing have been instrumental in addressing the issue of non-specificity and toxicity for therapeutic use. These application-based modifications may help in further increasing the efficiency of gene editing in protozoan parasites.
Collapse
Affiliation(s)
- Apurva Kirti
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Manish Sharma
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Komal Rani
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Abhisheka Bansal
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India.
| |
Collapse
|
21
|
Le Manach C, Dam J, Woodland JG, Kaur G, Khonde LP, Brunschwig C, Njoroge M, Wicht KJ, Horatscheck A, Paquet T, Boyle GA, Gibhard L, Taylor D, Lawrence N, Yeo T, Mok S, Eastman RT, Dorjsuren D, Talley DC, Guo H, Simeonov A, Reader J, van der Watt M, Erlank E, Venter N, Zawada JW, Aswat A, Nardini L, Coetzer TL, Lauterbach SB, Bezuidenhout BC, Theron A, Mancama D, Koekemoer LL, Birkholtz LM, Wittlin S, Delves M, Ottilie S, Winzeler EA, von Geldern TW, Smith D, Fidock DA, Street LJ, Basarab GS, Duffy J, Chibale K. Identification and Profiling of a Novel Diazaspiro[3.4]octane Chemical Series Active against Multiple Stages of the Human Malaria Parasite Plasmodium falciparum and Optimization Efforts. J Med Chem 2021; 64:2291-2309. [PMID: 33573376 DOI: 10.1021/acs.jmedchem.1c00034] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A novel diazaspiro[3.4]octane series was identified from a Plasmodium falciparum whole-cell high-throughput screening campaign. Hits displayed activity against multiple stages of the parasite lifecycle, which together with a novel sp3-rich scaffold provided an attractive starting point for a hit-to-lead medicinal chemistry optimization and biological profiling program. Structure-activity-relationship studies led to the identification of compounds that showed low nanomolar asexual blood-stage activity (<50 nM) together with strong gametocyte sterilizing properties that translated to transmission-blocking activity in the standard membrane feeding assay. Mechanistic studies through resistance selection with one of the analogues followed by whole-genome sequencing implicated the P. falciparum cyclic amine resistance locus in the mode of resistance.
Collapse
Affiliation(s)
- Claire Le Manach
- Drug Discovery and Development Center (H3D), University of Cape Town, Rondebosch 7701, South Africa
| | - Jean Dam
- Drug Discovery and Development Center (H3D), University of Cape Town, Rondebosch 7701, South Africa
| | - John G Woodland
- Drug Discovery and Development Center (H3D), University of Cape Town, Rondebosch 7701, South Africa
| | - Gurminder Kaur
- Drug Discovery and Development Center (H3D), University of Cape Town, Rondebosch 7701, South Africa
| | - Lutete P Khonde
- Drug Discovery and Development Center (H3D), University of Cape Town, Rondebosch 7701, South Africa
| | - Christel Brunschwig
- Drug Discovery and Development Center (H3D), Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Observatory 7925, South Africa
| | - Mathew Njoroge
- Drug Discovery and Development Center (H3D), Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Observatory 7925, South Africa
| | - Kathryn J Wicht
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, New York 10032, United States
| | - André Horatscheck
- Drug Discovery and Development Center (H3D), University of Cape Town, Rondebosch 7701, South Africa
| | - Tanya Paquet
- Drug Discovery and Development Center (H3D), University of Cape Town, Rondebosch 7701, South Africa
| | - Grant A Boyle
- Drug Discovery and Development Center (H3D), University of Cape Town, Rondebosch 7701, South Africa
| | - Liezl Gibhard
- Drug Discovery and Development Center (H3D), Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Observatory 7925, South Africa
| | - Dale Taylor
- Drug Discovery and Development Center (H3D), Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Observatory 7925, South Africa
| | - Nina Lawrence
- Drug Discovery and Development Center (H3D), Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Observatory 7925, South Africa
| | - 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
| | - Richard T Eastman
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Dorjbal Dorjsuren
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Daniel C Talley
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Hui Guo
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Anton Simeonov
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Janette Reader
- Department of Biochemistry, Genetics and Microbiology, Institute for Sustainable Malaria Control, University of Pretoria, Hatfield, Pretoria 0028, South Africa
| | - Mariëtte van der Watt
- Department of Biochemistry, Genetics and Microbiology, Institute for Sustainable Malaria Control, University of Pretoria, Hatfield, Pretoria 0028, South Africa
| | - Erica Erlank
- Wits Research Institute for Malaria, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa.,Centre for Emerging Zoonotic and Parasitic Diseases, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg 2192, South Africa
| | - Nelius Venter
- Wits Research Institute for Malaria, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa.,Centre for Emerging Zoonotic and Parasitic Diseases, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg 2192, South Africa
| | - Jacek W Zawada
- Wits Research Institute for Malaria, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa.,Centre for Emerging Zoonotic and Parasitic Diseases, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg 2192, South Africa
| | - Ayesha Aswat
- Wits Research Institute for Malaria, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa.,Centre for Emerging Zoonotic and Parasitic Diseases, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg 2192, South Africa
| | - Luisa Nardini
- Wits Research Institute for Malaria, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa.,Centre for Emerging Zoonotic and Parasitic Diseases, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg 2192, South Africa
| | - Theresa L Coetzer
- Wits Research Institute for Malaria, School of Pathology, Department of Molecular Medicine and Haematology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa
| | - Sonja B Lauterbach
- Wits Research Institute for Malaria, School of Pathology, Department of Molecular Medicine and Haematology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa
| | - Belinda C Bezuidenhout
- Wits Research Institute for Malaria, School of Pathology, Department of Molecular Medicine and Haematology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa
| | - Anjo Theron
- Biosciences, Council for Scientific and Industrial Research, P.O. Box 395, Pretoria 0001, South Africa
| | - Dalu Mancama
- Biosciences, Council for Scientific and Industrial Research, P.O. Box 395, Pretoria 0001, South Africa
| | - Lizette L Koekemoer
- Wits Research Institute for Malaria, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa.,Centre for Emerging Zoonotic and Parasitic Diseases, National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg 2192, South Africa
| | - Lyn-Marie Birkholtz
- Department of Biochemistry, Genetics and Microbiology, Institute for Sustainable Malaria Control, University of Pretoria, Hatfield, Pretoria 0028, South Africa
| | - Sergio Wittlin
- Swiss Tropical and Public Health Institute, Socinstrasse 57, 4002 Basel, Switzerland.,University of Basel, 4003 Basel, Switzerland
| | - Michael Delves
- Imperial College London, South Kensington, London SW7 2AZ, U.K.,London School of Hygiene and Tropical Medicine, London WC1E 7HT, U.K
| | - Sabine Ottilie
- Division of Host-Microbe Systems & Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, California 92093-076, United States
| | - Elizabeth A Winzeler
- Division of Host-Microbe Systems & Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, California 92093-076, United States
| | | | | | - 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
| | - Leslie J Street
- Drug Discovery and Development Center (H3D), University of Cape Town, Rondebosch 7701, South Africa
| | - Gregory S Basarab
- Drug Discovery and Development Center (H3D), University of Cape Town, Rondebosch 7701, South Africa
| | - James Duffy
- Medicines for Malaria Venture, ICC, Route de Pré-Bois 20, P.O. Box 1826, 1215 Geneva, Switzerland
| | - Kelly Chibale
- Drug Discovery and Development Center (H3D), University of Cape Town, Rondebosch 7701, South Africa.,South African Medical Research Council, Drug Discovery and Development Research Unit, Department of Chemistry and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa
| |
Collapse
|
22
|
Openshaw R, Maepa K, Benjamin SJ, Wainwright L, Combrinck JM, Hunter R, Egan TJ. A Diverse Range of Hemozoin Inhibiting Scaffolds Act on Plasmodium falciparum as Heme Complexes. ACS Infect Dis 2021; 7:362-376. [PMID: 33430579 DOI: 10.1021/acsinfecdis.0c00680] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
A diverse series of hemozoin-inhibiting quinolines, benzamides, triarylimidazoles, quinazolines, benzimidazoles, benzoxazoles, and benzothiazoles have been found to lead to exchangeable heme levels in cultured Plasmodium falciparum (NF54) that ranged over an order of magnitude at the IC50. Surprisingly, less active compounds often exhibited higher levels of exchangeable heme than more active ones. Quantities of intracellular inhibitor measured using the inoculum effect exhibited a linear correlation with exchangeable heme, suggesting formation of heme-inhibitor complexes in the parasite. In an effort to confirm this, the presence of a Br atom in one of the benzimidazole derivatives was exploited to image its distribution in the parasite using electron spectroscopic imaging of Br, an element not naturally abundant in cells. This showed that the compound colocalized with iron, consistent with its presence as a heme complex. Direct evidence for this complex was then obtained using confocal Raman microscopy. Exchangeable heme and inhibitor were found to increase with decreased rate of killing, suggesting that slow-acting compounds have more time to build up exchangeable heme complexes. Lastly, some but not all compounds evidently cause pro-oxidant effects because their activity could be attenuated with N-acetylcysteine and potentiated with t-butyl hydroperoxide. Collectively, these findings suggest that hemozoin inhibitors act as complexes with free heme, each with its own unique activity.
Collapse
Affiliation(s)
- Roxanne Openshaw
- Department of Chemistry, University of Cape Town, Private Bag Rondebosch, Cape Town 7701, South Africa
| | | | - Stefan J. Benjamin
- Department of Chemistry, University of Cape Town, Private Bag Rondebosch, Cape Town 7701, South Africa
| | - Lauren Wainwright
- Department of Chemistry, University of Cape Town, Private Bag Rondebosch, Cape Town 7701, South Africa
| | | | - Roger Hunter
- Department of Chemistry, University of Cape Town, Private Bag Rondebosch, Cape Town 7701, South Africa
| | - Timothy J. Egan
- Department of Chemistry, University of Cape Town, Private Bag Rondebosch, Cape Town 7701, South Africa
| |
Collapse
|
23
|
Patra KP, Kaur H, Kolli SK, Wozniak JM, Prieto JH, Yates JR, Gonzalez DJ, Janse CJ, Vinetz JM. A Hetero-Multimeric Chitinase-Containing Plasmodium falciparum and Plasmodium gallinaceum Ookinete-Secreted Protein Complex Involved in Mosquito Midgut Invasion. Front Cell Infect Microbiol 2021; 10:615343. [PMID: 33489941 PMCID: PMC7821095 DOI: 10.3389/fcimb.2020.615343] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 11/30/2020] [Indexed: 12/14/2022] Open
Abstract
Malaria parasites are transmitted by Anopheles mosquitoes. During its life cycle in the mosquito vector the Plasmodium ookinete escapes the proteolytic milieu of the post-blood meal midgut by traversing the midgut wall. This process requires penetration of the chitin-containing peritrophic matrix lining the midgut epithelium, which depends in part on ookinete-secreted chitinases. Plasmodium falciparum ookinetes have one chitinase (PfCHT1), whereas ookinetes of the avian-infecting parasite, P. gallinaceum, have two, a long and a short form, PgCHT1 and PgCHT2, respectively. Published data indicates that PgCHT2 forms a high molecular weight (HMW) reduction-sensitive complex; and one binding partner is the ookinete-produced von Willebrand A-domain-containing protein, WARP. Size exclusion chromatography data reported here show that P. gallinaceum PgCHT2 and its ortholog, P. falciparum PfCHT1 are covalently-linked components of a HMW chitinase-containing complex (> 1,300 kDa). Mass spectrometry of ookinete-secreted proteins isolated using a new chitin bead pull-down method identified chitinase-associated proteins in P. falciparum and P. gallinaceum ookinete-conditioned culture media. Mass spectrometry of this complex showed the presence of several micronemal proteins including von Willebrand factor A domain-related protein (WARP), ookinete surface enolase, and secreted ookinete adhesive protein (SOAP). To test the hypothesis that ookinete-produced PfCHT1 can form a high molecular homo-multimer or, alternatively, interacts with P. berghei ookinete-produced proteins to produce an HMW hetero-multimer, we created chimeric P. berghei parasites expressing PfCHT1 to replace PbCHT1, enabling the production of large numbers of PfCHT1-expressing ookinetes. We show that chimeric P. berghei ookinetes express monomeric PfCHT1, but a HMW complex containing PfCHT1 is not present. A better understanding of the chitinase-containing HMW complex may enhance development of next-generation vaccines or drugs that target malaria transmission stages.
Collapse
Affiliation(s)
- Kailash P Patra
- Section of Infectious Diseases, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, United States
| | - Hargobinder Kaur
- Section of Infectious Diseases, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, United States
| | - Surendra Kumar Kolli
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Jacob M Wozniak
- Department of Pharmacology and the Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, United States
| | - Judith Helena Prieto
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, United States.,Department of Chemistry, Western Connecticut State University, Danbury, CT, United States
| | - John R Yates
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, United States
| | - David J Gonzalez
- Department of Pharmacology and the Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, United States
| | - Chris J Janse
- Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Joseph M Vinetz
- Section of Infectious Diseases, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, United States
| |
Collapse
|
24
|
Yi XG, Lai FP, Yan YY, Zhang C, Li WP. Preparation, crystal structures, properties, and time-dependent density functional theory of two cobalt complexes with 3-hydroxy-2-methyl-quinoline-4-carboxylate. JOURNAL OF CHEMICAL RESEARCH 2020. [DOI: 10.1177/1747519820948363] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
One organic compound [HMCA]2 (1) and two novel cobalt complexes [Co(MCA)(bipy)(H2O)]·(H2O) (2), [Co2(MCA)2(Phen)3]·3(H2O) (3) are synthesized by a solvothermal approach and are structurally determined by single-crystal X-ray diffraction. Compound 1 exhibits a two-dimensional structure by hydrogen bond and π. . .π stacking interaction. The complexes exhibit a three-dimensional and one-dimensional metal-organic framework. Solid-state photoluminescence spectrums reveal that they show blue emission bands at 449 nm, and theoretical calculation results of time-dependent density functional theory show that all belong to ligand-to-ligand charge transfer. Solid-state diffuse reflectance spectrums reveal the presence of a narrow optical band gap of 1.72 and 1.60 eV in the complexes 2 and 3, respectively.
Collapse
Affiliation(s)
- Xiu-Guang Yi
- School of Chemistry and Chemical Engineering, Jinggangshan University, Ji’an, P.R. China
| | - Fei-Ping Lai
- School of Chemistry and Chemical Engineering, Jinggangshan University, Ji’an, P.R. China
| | - Yun-Yi Yan
- School of Chemistry and Chemical Engineering, Jinggangshan University, Ji’an, P.R. China
| | - Cong Zhang
- School of Chemistry and Chemical Engineering, Jinggangshan University, Ji’an, P.R. China
| | - Wen-Ping Li
- School of Chemistry and Chemical Engineering, Jinggangshan University, Ji’an, P.R. China
| |
Collapse
|
25
|
de Souza Pereira C, Costa Quadros H, Magalhaes Moreira DR, Castro W, Santos De Deus Da Silva RI, Botelho Pereira Soares M, Fontinha D, Prudêncio M, Schmitz V, Dos Santos HF, Gendrot M, Fonta I, Mosnier J, Pradines B, Navarro M. A Novel Hybrid of Chloroquine and Primaquine Linked by Gold(I): Multitarget and Multiphase Antiplasmodial Agent. ChemMedChem 2020; 16:662-678. [PMID: 33231370 DOI: 10.1002/cmdc.202000653] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 11/02/2020] [Indexed: 12/23/2022]
Abstract
Plasmodium parasites kill 435 000 people around the world every year due to unavailable vaccines, a limited arsenal of antimalarial drugs, delayed treatment, and the reduced clinical effectiveness of current practices caused by drug resistance. Therefore, there is an urgent need to discover and develop new antiplasmodial candidates. In this work, we present a novel strategy to develop a multitarget metallic hybrid antimalarial agent with possible dual efficacy in both sexual and asexual erythrocytic stages. A hybrid of antimalarial drugs (chloroquine and primaquine) linked by gold(I) was synthesized and characterized by spectroscopic and analytical techniques. The CQPQ-gold(I) hybrid molecule affects essential parasite targets, it inhibits β-hematin formation and interacts moderately with the DNA minor groove. Its interaction with PfTrxR was also examined in computational modeling studies. The CQPQ-gold(I) hybrid displayed an excellent in vitro antimalarial activity against the blood-stage of Plasmodium falciparum and liver-stage of Plasmodium berghei and efficacy in vivo against P. berghei, thereby demonstrating its multiple-stage antiplasmodial activity. This metallic hybrid is a promising chemotherapeutic agent that could act in the treatment, prevention, and transmission of malaria.
Collapse
Affiliation(s)
- Caroline de Souza Pereira
- Departamento de Química, Universidade Federal de Juiz de Fora, Rua José Lourenço Kelmer, s/n - Campus Universitário, Bairro Martelos, CEP 36036-900, Juiz de Fora, Minas Gerais, Brasil
| | - Helenita Costa Quadros
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Av. Waldemar Falcão, 121, Candeal, Salvador, Bahia, Brasil
| | | | - William Castro
- Centro de Química, Instituto Venezolano de Investigaciones Científicas (IVIC), Centro de Química, Carretera Panamericana, Km 11, Altos de Pipe, San Antonio de los Altos Miranda, 1020-A, Caracas, Venezuela
| | | | | | - Diana Fontinha
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa <, Lisboa, Portugal
| | - Miguel Prudêncio
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa <, Lisboa, Portugal
| | - Vinicius Schmitz
- Departamento de Química, Universidade Federal de Juiz de Fora, Rua José Lourenço Kelmer, s/n - Campus Universitário, Bairro Martelos, CEP 36036-900, Juiz de Fora, Minas Gerais, Brasil
| | - Hélio F Dos Santos
- Departamento de Química, Universidade Federal de Juiz de Fora, Rua José Lourenço Kelmer, s/n - Campus Universitário, Bairro Martelos, CEP 36036-900, Juiz de Fora, Minas Gerais, Brasil
| | - Mathieu Gendrot
- Unité Parasitologie et entomologie, Institut de recherche biomédicale des armées, 19-21 Bd Jean Moulin, 13005, Marseille, France.,Aix-Marseille Univ, IRD, SSA, AP-HM, VITROME, 19-21 Bd Jean Moulin, 13005, Marseille, France.,IHU Méditerranée Infection, 19-21 Bd Jean Moulin, 13005, Marseille, France
| | - Isabelle Fonta
- Unité Parasitologie et entomologie, Institut de recherche biomédicale des armées, 19-21 Bd Jean Moulin, 13005, Marseille, France.,Aix-Marseille Univ, IRD, SSA, AP-HM, VITROME, 19-21 Bd Jean Moulin, 13005, Marseille, France.,IHU Méditerranée Infection, 19-21 Bd Jean Moulin, 13005, Marseille, France.,Centre National de Référence du Paludisme, 19-21 Bd Jean Moulin, 13005, Marseille, France
| | - Joel Mosnier
- Unité Parasitologie et entomologie, Institut de recherche biomédicale des armées, 19-21 Bd Jean Moulin, 13005, Marseille, France.,Aix-Marseille Univ, IRD, SSA, AP-HM, VITROME, 19-21 Bd Jean Moulin, 13005, Marseille, France.,IHU Méditerranée Infection, 19-21 Bd Jean Moulin, 13005, Marseille, France.,Centre National de Référence du Paludisme, 19-21 Bd Jean Moulin, 13005, Marseille, France
| | - Bruno Pradines
- Unité Parasitologie et entomologie, Institut de recherche biomédicale des armées, 19-21 Bd Jean Moulin, 13005, Marseille, France.,Aix-Marseille Univ, IRD, SSA, AP-HM, VITROME, 19-21 Bd Jean Moulin, 13005, Marseille, France.,IHU Méditerranée Infection, 19-21 Bd Jean Moulin, 13005, Marseille, France.,Centre National de Référence du Paludisme, 19-21 Bd Jean Moulin, 13005, Marseille, France
| | - Maribel Navarro
- Departamento de Química, Universidade Federal de Juiz de Fora, Rua José Lourenço Kelmer, s/n - Campus Universitário, Bairro Martelos, CEP 36036-900, Juiz de Fora, Minas Gerais, Brasil
| |
Collapse
|
26
|
Veale CGL, Müller R. Recent Highlights in Anti-infective Medicinal Chemistry from South Africa. ChemMedChem 2020; 15:809-826. [PMID: 32149446 DOI: 10.1002/cmdc.202000086] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Indexed: 12/17/2022]
Abstract
Global advancements in biological technologies have vastly increased the variety of and accessibility to bioassay platforms, while simultaneously improving our understanding of druggable chemical space. In the South African context, this has resulted in a rapid expansion in the number of medicinal chemistry programmes currently operating, particularly on university campuses. Furthermore, the modern medicinal chemist has the advantage of being able to incorporate data from numerous related disciplines into the medicinal chemistry process, allowing for informed molecular design to play a far greater role than previously possible. Accordingly, this review focusses on recent highlights in drug-discovery programmes, in which South African medicinal chemistry groups have played a substantive role in the design and optimisation of biologically active compounds which contribute to the search for promising agents for infectious disease.
Collapse
Affiliation(s)
- Clinton G L Veale
- School of Chemistry and Physics, Pietermaritzburg Campus, University of KwaZulu-Natal, Private Bag X01, Scottsville, 3209, South Africa
| | - Ronel Müller
- School of Chemistry and Physics, Pietermaritzburg Campus, University of KwaZulu-Natal, Private Bag X01, Scottsville, 3209, South Africa
| |
Collapse
|
27
|
Murithi JM, Owen ES, Istvan ES, Lee MCS, Ottilie S, Chibale K, Goldberg DE, Winzeler EA, Llinás M, Fidock DA, Vanaerschot M. Combining Stage Specificity and Metabolomic Profiling to Advance Antimalarial Drug Discovery. Cell Chem Biol 2019; 27:158-171.e3. [PMID: 31813848 PMCID: PMC7031696 DOI: 10.1016/j.chembiol.2019.11.009] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 10/18/2019] [Accepted: 11/14/2019] [Indexed: 01/08/2023]
Abstract
We report detailed susceptibility profiling of asexual blood stages of the malaria parasite Plasmodium falciparum to clinical and experimental antimalarials, combined with metabolomic fingerprinting. Results revealed a variety of stage-specific and metabolic profiles that differentiated the modes of action of clinical antimalarials including chloroquine, piperaquine, lumefantrine, and mefloquine, and identified late trophozoite-specific peak activity and stage-specific biphasic dose-responses for the mitochondrial inhibitors DSM265 and atovaquone. We also identified experimental antimalarials hitting previously unexplored druggable pathways as reflected by their unique stage specificity and/or metabolic profiles. These included several ring-active compounds, ones affecting hemoglobin catabolism through distinct pathways, and mitochondrial inhibitors with lower propensities for resistance than either DSM265 or atovaquone. This approach, also applicable to other microbes that undergo multiple differentiation steps, provides an effective tool to prioritize compounds for further development within the context of combination therapies.
Collapse
Affiliation(s)
- James M Murithi
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Edward S Owen
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
| | - Eva S Istvan
- Department of Medicine, Division of Infectious Diseases, and Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis MO 63130, USA
| | - Marcus C S Lee
- Parasites and Microbes Programme, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire CB10 1SA, UK
| | - Sabine Ottilie
- School of Medicine, University of California San Diego (UCSD), La Jolla, CA 92093, USA
| | - Kelly Chibale
- Drug Discovery and Development Centre (H3D), University of Cape Town, Rondebosch 7701, South Africa; South African Medical Research Council Drug Discovery and Development Research Unit, Department of Chemistry & Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa
| | - Daniel E Goldberg
- Department of Medicine, Division of Infectious Diseases, and Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis MO 63130, USA
| | - Elizabeth A Winzeler
- School of Medicine, University of California San Diego (UCSD), La Jolla, CA 92093, USA
| | - Manuel Llinás
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA; Department of Chemistry, Pennsylvania State University, University Park, PA 16802, USA; Huck Center for Malaria Research, Pennsylvania State University, University Park, PA 16802, USA
| | - David A Fidock
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA; Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA.
| | - Manu Vanaerschot
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA.
| |
Collapse
|
28
|
Martin RE. The transportome of the malaria parasite. Biol Rev Camb Philos Soc 2019; 95:305-332. [PMID: 31701663 DOI: 10.1111/brv.12565] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 10/02/2019] [Accepted: 10/04/2019] [Indexed: 12/15/2022]
Abstract
Membrane transport proteins, also known as transporters, control the movement of ions, nutrients, metabolites, and waste products across the membranes of a cell and are central to its biology. Proteins of this type also serve as drug targets and are key players in the phenomenon of drug resistance. The malaria parasite has a relatively reduced transportome, with only approximately 2.5% of its genes encoding transporters. Even so, assigning functions and physiological roles to these proteins, and ascertaining their contributions to drug action and drug resistance, has been very challenging. This review presents a detailed critique and synthesis of the disruption phenotypes, protein subcellular localisations, protein functions (observed or predicted), and links to antimalarial drug resistance for each of the parasite's transporter genes. The breadth and depth of the gene disruption data are particularly impressive, with at least one phenotype determined in the parasite's asexual blood stage for each transporter gene, and multiple phenotypes available for 76% of the genes. Analysis of the curated data set revealed there to be relatively little redundancy in the Plasmodium transportome; almost two-thirds of the parasite's transporter genes are essential or required for normal growth in the asexual blood stage of the parasite, and this proportion increased to 78% when the disruption phenotypes available for the other parasite life stages were included in the analysis. These observations, together with the finding that 22% of the transportome is implicated in the parasite's resistance to existing antimalarials and/or drugs within the development pipeline, indicate that transporters are likely to serve, or are already serving, as drug targets. Integration of the different biological and bioinformatic data sets also enabled the selection of candidates for transport processes known to be essential for parasite survival, but for which the underlying proteins have thus far remained undiscovered. These include potential transporters of pantothenate, isoleucine, or isopentenyl diphosphate, as well as putative anion-selective channels that may serve as the pore component of the parasite's 'new permeation pathways'. Other novel insights into the parasite's biology included the identification of transporters for the potential development of antimalarial treatments, transmission-blocking drugs, prophylactics, and genetically attenuated vaccines. The syntheses presented herein set a foundation for elucidating the functions and physiological roles of key members of the Plasmodium transportome and, ultimately, to explore and realise their potential as therapeutic targets.
Collapse
Affiliation(s)
- Rowena E Martin
- Research School of Biology, Australian National University, Canberra, Australian Capital Territory, Australia
| |
Collapse
|
29
|
Lee MCS, Lindner SE, Lopez-Rubio JJ, Llinás M. Cutting back malaria: CRISPR/Cas9 genome editing of Plasmodium. Brief Funct Genomics 2019; 18:281-289. [PMID: 31365053 PMCID: PMC6859820 DOI: 10.1093/bfgp/elz012] [Citation(s) in RCA: 37] [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: 12/19/2018] [Revised: 04/15/2019] [Accepted: 05/20/2019] [Indexed: 12/15/2022] Open
Abstract
CRISPR/Cas9 approaches are revolutionizing our ability to perform functional genomics across a wide range of organisms, including the Plasmodium parasites that cause malaria. The ability to deliver single point mutations, epitope tags and gene deletions at increased speed and scale is enabling our understanding of the biology of these complex parasites, and pointing to potential new therapeutic targets. In this review, we describe some of the biological and technical considerations for designing CRISPR-based experiments, and discuss potential future developments that broaden the applications for CRISPR/Cas9 interrogation of the malaria parasite genome.
Collapse
Affiliation(s)
- Marcus C S Lee
- Parasites and Microbes Programme, Wellcome Sanger Institute, Hinxton, UK
| | - Scott E Lindner
- Department of Biochemistry and Molecular Biology, Huck Center for Malaria Research, The Pennsylvania State University, Pennsylvania, USA
| | - Jose-Juan Lopez-Rubio
- Dynamique des Interactions Membranaires Normales et Pathologiques, UMR5235 CNRS, INSERM, Université Montpellier, Montpellier, France
| | - Manuel Llinás
- Department of Biochemistry and Molecular Biology, Huck Center for Malaria Research, The Pennsylvania State University, Pennsylvania, USA
- Department of Chemistry, The Pennsylvania State University, Pennsylvania, USA
| |
Collapse
|
30
|
Novel magnetic nanoparticles with morpholine tags as multirole catalyst for synthesis of hexahydroquinolines and 2-amino-4,6-diphenylnicotinonitriles through vinylogous anomeric-based oxidation. RESEARCH ON CHEMICAL INTERMEDIATES 2019. [DOI: 10.1007/s11164-019-03802-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
31
|
Ng CL, Fidock DA. Plasmodium falciparum In Vitro Drug Resistance Selections and Gene Editing. Methods Mol Biol 2019; 2013:123-140. [PMID: 31267498 DOI: 10.1007/978-1-4939-9550-9_9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Malaria continues to be a global health burden, threatening over 40% of the world's population. Drug resistance in Plasmodium falciparum, the etiological agent of the majority of human malaria cases, is compromising elimination efforts. New approaches to treating drug-resistant malaria benefit from defining resistance liabilities of known antimalarial agents and compounds in development and defining genetic changes that mediate loss of parasite susceptibility. Here, we present protocols for in vitro selection of drug-resistant parasites and for site-directed gene editing of candidate resistance mediators to test for causality.
Collapse
Affiliation(s)
- Caroline L Ng
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - David A Fidock
- Department of Microbiology and Immunology, 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
|
32
|
Delpe Acharige AMDS, Brennan MPC, Lauder K, McMahon F, Odebunmi AO, Durrant MC. Computational insights into the inhibition of β-haematin crystallization by antimalarial drugs. Dalton Trans 2018; 47:15364-15381. [PMID: 30298161 DOI: 10.1039/c8dt03369b] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
During the red blood cell phase of their life cycle, malaria parasites digest their host's haemoglobin, with concomitant release of potentially toxic iron(iii) protoporphyrin IX (FePPIX). The parasites' strategy for detoxification of FePPIX involves its crystallization to haemozoin, such that the build-up of free haem in solution is avoided. Antimalarial drugs of both historical importance and current clinical use are known to be capable of disrupting the growth of crystals of β-haematin, which is the synthetic equivalent of haemozoin. Hence, the disruption of haemozoin crystal growth is implicated as a possible mode of action of such drugs. However, the details of β-haematin crystal poisoning at the molecular level have yet to be fully elucidated. In this study, we have used a combination of density functional theory (DFT) and molecular modelling to examine the possible modes of action of ten different antimalarial drugs, including quinine-type aliphatic alcohols, amodiaquine-type phenols, and chloroquine-type aliphatic diamines. The DFT calculations indicate that each of the drugs can form at least one molecular complex with FePPIX. These complexes have 1 : 1 or 2 : 1 FePPIX : drug stoichiometries and all of them incorporate Fe-O bonds, formed either by direct coordination of a zwitterionic form of the drug, or by deprotonation of water. Most of the drugs can form more than one such complex. We have used the DFT model structures to explore the possible formation of a monolayer of each drug-haem complex on four of the β-haematin crystal faces. In all cases, the drug complexes can form a monolayer on the fast-growing {001} and {011} faces, but not on the slower growing {010} and {100} faces. Additional modelling of the chloroquine and quinidine complexes shows that individual molecules of these species can also obstruct the growth of new layers on other crystal faces. The implications of these observations for antimalarial drug development are discussed.
Collapse
Affiliation(s)
- Anjana M D S Delpe Acharige
- Faculty of Health and Life Sciences, Northumbria University, Ellison Building, Newcastle-upon-Tyne NE2 8ST, UK.
| | | | | | | | | | | |
Collapse
|
33
|
5-Oxo-hexahydroquinoline: an attractive scaffold with diverse biological activities. Mol Divers 2018; 23:471-508. [PMID: 30390186 DOI: 10.1007/s11030-018-9886-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 10/17/2018] [Indexed: 12/31/2022]
Abstract
5-Oxo-hexahydroquinoline (5-oxo-HHQ) represents a biologically attractive fused heterocyclic core. Various synthetic analogs of 5-oxo-HHQ have been synthesized and assessed for different biological activities. Some derivatives have exhibited myorelaxant, analgesic, anticancer, antibacterial, antifungal, antitubercular, antimalarial, antioxidant, anti-inflammatory, multidrug resistance reversal, anti-Alzheimer, neuroprotective, antidiabetic, antidyslipidemic and antiosteoporotic activities. This review provides a comprehensive report regarding the preparation and pharmacological characterization of 5-oxo-HHQ derivatives that have been reported so far. This information will be beneficial for medicinal chemists in the field of drug discovery to design and develop new and potent therapeutical agents bearing the 5-oxo-HHQ nucleus.
Collapse
|
34
|
Pasinszki T, Krebsz M, Lajgut GG, Kocsis T, Kótai L, Kauthale S, Tekale S, Pawar R. Copper nanoparticles grafted on carbon microspheres as novel heterogeneous catalysts and their application for the reduction of nitrophenol and one-pot multicomponent synthesis of hexahydroquinolines. NEW J CHEM 2018. [DOI: 10.1039/c7nj03562d] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Carbon microsphere-supported Cu nanoparticles were fabricated, characterized, and applied for synthesis.
Collapse
Affiliation(s)
- Tibor Pasinszki
- ELTE Eötvös Loránd University
- Institute of Chemistry
- Budapest
- Hungary
| | - Melinda Krebsz
- ELTE Eötvös Loránd University
- Institute of Chemistry
- Budapest
- Hungary
| | | | - Tünde Kocsis
- Research Centre for Natural Sciences
- Hungarian Academy of Sciences
- Budapest
- Hungary
| | - László Kótai
- Research Centre for Natural Sciences
- Hungarian Academy of Sciences
- Budapest
- Hungary
| | | | - Sunil Tekale
- Department of Chemistry
- Deogiri College
- Aurangabad (MS)
- India
| | - Rajendra Pawar
- Department of Chemistry
- Deogiri College
- Aurangabad (MS)
- India
| |
Collapse
|
35
|
Crunkhorn S. Blocking malaria transmission. Nat Rev Drug Discov 2017; 16:680. [DOI: 10.1038/nrd.2017.184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|