1
|
Early AM, Pelleau S, Musset L, Neafsey DE. Temporal Patterns of Haplotypic and Allelic Diversity Reflect the Changing Selection Landscape of the Malaria Parasite Plasmodium falciparum. Mol Biol Evol 2025; 42:msaf075. [PMID: 40164958 PMCID: PMC12004115 DOI: 10.1093/molbev/msaf075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 03/14/2025] [Accepted: 03/21/2025] [Indexed: 04/02/2025] Open
Abstract
The malaria parasite Plasmodium falciparum regularly confronts orchestrated changes in frontline drug treatment that drastically alter its selection landscape. When this has occurred, the parasite has successfully adapted to new drugs through novel resistance mutations. These novel mutations, however, emerge in a genetic background already shaped by prior drug selection. In some instances, selection imposed by different drugs targets the same loci in either synergistic or antagonistic ways, which may leave genomic signatures that are hard to attribute to a specific agent. Here, we use two approaches for detecting sequential bouts of drug adaptation: haplotype-based selection testing and temporal changes in allele frequencies. Using a set of longitudinal samples from French Guiana, we determine that since the official introduction of artemisinin combination therapy in 2007 there have been rapid hard selective sweeps at both known and novel loci. At four high-profile genes with demonstrated involvement in drug resistance (pfcrt, pfmdr1, pfaat1, and pfgch1), we see selection signals both before and after drug regime change; however, selection favored different haplotypes in the two time periods. Similarly, allele frequency analysis identified coding variants whose frequency trajectory changed signs under the new drug pressure. These selected alleles were enriched for genes implicated in artemisinin or partner-drug resistance in other global populations. Overall, these results suggest that drug resistance in P. falciparum is governed by known alleles of large effect along with a polygenic architecture of potentially more subtle variants, any of which can experience fitness reversals under distinct drug regimes.
Collapse
Affiliation(s)
- Angela M Early
- Infectious Disease and Microbiome Program, Broad Institute, Cambridge, MA 02142, USA
| | - Stéphane Pelleau
- Infectious Diseases Epidemiology and Analytics Unit, Department of Global Health, lnstitut Pasteur, Université Paris Cité, Paris 75015, France
- Centre National de Référence du Paludisme, World Health Organization Collaborating Center for Surveillance of Antimalarial Drug Resistance, lnstitut Pasteur de la Guyane, Cayenne 97300, French Guiana
| | - Lise Musset
- Centre National de Référence du Paludisme, World Health Organization Collaborating Center for Surveillance of Antimalarial Drug Resistance, lnstitut Pasteur de la Guyane, Cayenne 97300, French Guiana
| | - Daniel E Neafsey
- Infectious Disease and Microbiome Program, Broad Institute, Cambridge, MA 02142, USA
- Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Harvard University, Boston, MA 02115, USA
| |
Collapse
|
2
|
Muriithi B, Chepngetich J, Gachie B, Thiong'o K, Gathirwa J, Kimani F, Mwitari P, Kiboi D. Structural and functional implications of MIT2 and NT2 mutations in amodiaquine and piperaquine resistant Plasmodium berghei parasites. Exp Parasitol 2025; 271:108923. [PMID: 40032183 DOI: 10.1016/j.exppara.2025.108923] [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: 06/04/2024] [Revised: 11/27/2024] [Accepted: 02/28/2025] [Indexed: 03/05/2025]
Abstract
Long-acting drugs, amodiaquine (AQ), lumefantrine (LM), and piperaquine (PQ), are vital components of artemisinin-based combination therapies (ACTs) for malaria treatment. However, the emergence of partial artemisinin-resistant parasites poses significant challenges, particularly in malaria-endemic regions. Despite extensive research, parasite's resistance mechanisms to these drugs still need complete elucidation. This study investigated the genetic basis of resistance to AQ, LM, and PQ using Plasmodium berghei, focusing on selected genes encoding transport proteins in Plasmodium species. In silico bioinformatics tools were used to map genes encoding transport proteins, their ligand-binding sites, and their conservation across different Plasmodium species. PCR amplification and sequence analysis were employed to examine single nucleotide polymorphisms (SNPs) in the genes encoding the selected transporters in AQ, LM, and PQ-resistant P. berghei. The structural impacts of the mutations were evaluated using AlphaFold, ITASSER, UCSF Chimera, and MOTIF Finder. Genes encoding CorA-like Mg2+ transporter protein (MIT2), nucleoside transporter 2 (NT2), ABC Transporter G family member 2 (ABCG2), and novel putative transporter 1 (NPT1) transport proteins with notable conserved motifs and ligand-binding motifs in Plasmodium species were selected and examined. In AQ-resistant (AQR) parasites, a non-synonymous mutation (I433∗) was found in MIT2. PQ-resistant (PQR) parasites possessed a non-synonymous mutation (D511H) in NT2 and a silent mutation in the NPT1 protein. No mutations were observed in the targeted regions of the transporters in LM-resistant (LMR) parasites, nor in the ligand-binding motifs of ABCG2 across all resistant strains. These findings suggest that selection pressure from AQ and PQ leads to mutations in MIT2 and NT2. Further investigation is required to understand how these mutations affect drug susceptibility on a functional level.
Collapse
Affiliation(s)
- Brenda Muriithi
- Centre for Biotechnology Research and Development, Kenya Medical Research Institute, P.O. Box 54840, 00200 Nairobi, Kenya; Department of Biochemistry, Jomo Kenyatta University of Agriculture and Technology, P.O. Box 62000, 00200 Nairobi, Kenya
| | - Jean Chepngetich
- Centre for Biotechnology Research and Development, Kenya Medical Research Institute, P.O. Box 54840, 00200 Nairobi, Kenya; Department of Biochemistry, Jomo Kenyatta University of Agriculture and Technology, P.O. Box 62000, 00200 Nairobi, Kenya; PanAfrican University Institute for Basic Sciences, Technology and Innovation is 62000, 00200, Nairobi, Kenya
| | - Beatrice Gachie
- Centre for Biotechnology Research and Development, Kenya Medical Research Institute, P.O. Box 54840, 00200 Nairobi, Kenya; PanAfrican University Institute for Basic Sciences, Technology and Innovation is 62000, 00200, Nairobi, Kenya
| | - Kevin Thiong'o
- Centre for Biotechnology Research and Development, Kenya Medical Research Institute, P.O. Box 54840, 00200 Nairobi, Kenya
| | - Jeremiah Gathirwa
- Centre for Traditional Medicine and Drug Research, Kenya Medical Research Institute, 54840, 00200, Nairobi, Kenya
| | - Francis Kimani
- Centre for Biotechnology Research and Development, Kenya Medical Research Institute, P.O. Box 54840, 00200 Nairobi, Kenya
| | - Peter Mwitari
- Centre for Traditional Medicine and Drug Research, Kenya Medical Research Institute, 54840, 00200, Nairobi, Kenya
| | - Daniel Kiboi
- Department of Biochemistry, Jomo Kenyatta University of Agriculture and Technology, P.O. Box 62000, 00200 Nairobi, Kenya.
| |
Collapse
|
3
|
Tisnerat C, Schneider J, Mustière R, Herrero A, Momha R, Damiani C, Agnamey P, Totet A, Marchivie M, Guillon J, Dassonville‐Klimpt A, Sonnet P. Synthesis of New Enantiopure Aminoalcohol Fluorenes as Promising Antimalarial Compounds. ChemMedChem 2025; 20:e202400790. [PMID: 39668712 PMCID: PMC11911300 DOI: 10.1002/cmdc.202400790] [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: 10/03/2024] [Revised: 12/06/2024] [Accepted: 12/06/2024] [Indexed: 12/14/2024]
Abstract
Herein, we report the design, synthesis, and characterisation of a new library of enantiopure aminoalcohol fluorenes, as well as their in vitro evaluation for biological properties, including activity against two strains of P. falciparum (3D7 and W2) and cytotoxicity on the HepG2 cell line. All tested compounds exhibited good to excellent antimalarial potency with IC50 values ranging from 0.7 to 70.2 nM whatever the strain. Interestingly, most compounds showed equal or better antimalarial activity compared to the reference drugs lumefantrine, mefloquine and chloroquine. Despite moderate cytotoxicity in the micromolar range, all aminoalcohol fluorenes displayed an excellent selectivity index higher than 100 due to strong antimalarial activity. Furthermore, we report in silico analyses of physicochemical and pharmacokinetic properties for all compounds, highlighting the drug-likeness of compound 10 and its promising potential for further studies.
Collapse
Affiliation(s)
| | | | - Romain Mustière
- UFR de PharmacieUniversité de Picardie Jules VerneAmiensFrance
| | - Aurélie Herrero
- UFR de PharmacieUniversité de Picardie Jules VerneAmiensFrance
| | - René Momha
- UFR de PharmacieUniversité de Picardie Jules VerneAmiensFrance
| | - Céline Damiani
- UFR de PharmacieUniversité de Picardie Jules VerneAmiensFrance
| | - Patrice Agnamey
- UFR de PharmacieUniversité de Picardie Jules VerneAmiensFrance
| | - Anne Totet
- UFR de PharmacieUniversité de Picardie Jules VerneAmiensFrance
| | - Mathieu Marchivie
- Université de BordeauxCNRSINPICMCBUMR 5026, PessacBordeauxF-33600France
| | - Jean Guillon
- INSERM U1212UMR CNRS 5320Laboratoire ARNAUFR des Sciences PharmaceutiquesUniversité de BordeauxBordeauxFrance
| | | | - Pascal Sonnet
- UFR de PharmacieUniversité de Picardie Jules VerneAmiensFrance
| |
Collapse
|
4
|
Bremers E, Butler JH, Do Amaral LS, Merino EF, Almolhim H, Zhou B, Baptista RP, Totrov M, Carlier PR, Cassera MB. Stereospecific Resistance to N2-Acyl Tetrahydro-β-carboline Antimalarials Is Mediated by a PfMDR1 Mutation That Confers Collateral Drug Sensitivity. ACS Infect Dis 2025; 11:529-542. [PMID: 39808111 PMCID: PMC11828674 DOI: 10.1021/acsinfecdis.4c01001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 12/18/2024] [Accepted: 12/27/2024] [Indexed: 01/16/2025]
Abstract
Half the world's population is at risk of developing a malaria infection, which is caused by parasites of the genus Plasmodium. Currently, resistance has been identified to all clinically available antimalarials, highlighting an urgent need to develop novel compounds and better understand common mechanisms of resistance. We previously identified a novel tetrahydro-β-carboline compound, PRC1590, which potently kills the malaria parasite. To better understand its mechanism of action, we selected for and characterized resistance to PRC1590 in Plasmodium falciparum. Through in vitro selection of resistance to PRC1590, we have identified that a single-nucleotide polymorphism on the parasite's multidrug resistance protein 1 (PfMDR1 G293V) mediates resistance to PRC1590. This mutation results in stereospecific resistance and sensitizes parasites to other antimalarials, such as mefloquine, quinine, and MMV019017. Intraerythrocytic asexual stage specificity assays have revealed that PRC1590 is most potent during the trophozoite stage when the parasite forms a single digestive vacuole (DV) and actively digests hemoglobin. Moreover, fluorescence microscopy revealed that PRC1590 disrupts the function of the DV, indicating a potential molecular target associated with this organelle. Our findings mark a significant step in understanding the mechanism of resistance and the mode of action of this emerging class of antimalarials. In addition, our results suggest a potential link between resistance mediated by PfMDR1 and PRC1590's molecular target. This research underscores the pressing need for future research aimed at investigating the intricate relationship between a compound's chemical scaffold, molecular target, and resistance mutations associated with PfMDR1.
Collapse
Affiliation(s)
- Emily
K. Bremers
- Department
of Biochemistry and Molecular Biology, University
of Georgia, Athens, Georgia 30602, United States
- Center
for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia 30602, United States
| | - Joshua H. Butler
- Department
of Biochemistry and Molecular Biology, University
of Georgia, Athens, Georgia 30602, United States
- Center
for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia 30602, United States
| | - Leticia S. Do Amaral
- Department
of Biochemistry and Molecular Biology, University
of Georgia, Athens, Georgia 30602, United States
- Center
for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia 30602, United States
| | - Emilio F. Merino
- Department
of Biochemistry and Molecular Biology, University
of Georgia, Athens, Georgia 30602, United States
- Center
for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia 30602, United States
| | - Hanan Almolhim
- Department
of Chemistry, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Bo Zhou
- Department
of Pharmaceutical Sciences, University of
Illinois Chicago, Chicago, Illinois 60612, United States
| | - Rodrigo P. Baptista
- Department
of Medicine, Houston Methodist Research
Institute, Houston, Texas 77030, United States
| | - Maxim Totrov
- MolSoft
LLC, San Diego, California 92121, United States
| | - Paul R. Carlier
- Department
of Chemistry, Virginia Tech, Blacksburg, Virginia 24061, United States
- Department
of Pharmaceutical Sciences, University of
Illinois Chicago, Chicago, Illinois 60612, United States
| | - Maria Belen Cassera
- Department
of Biochemistry and Molecular Biology, University
of Georgia, Athens, Georgia 30602, United States
- Center
for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia 30602, United States
| |
Collapse
|
5
|
Schäfer TM, Pessanha de Carvalho L, Inoue J, Kreidenweiss A, Held J. The problem of antimalarial resistance and its implications for drug discovery. Expert Opin Drug Discov 2024; 19:209-224. [PMID: 38108082 DOI: 10.1080/17460441.2023.2284820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 11/14/2023] [Indexed: 12/19/2023]
Abstract
INTRODUCTION Malaria remains a devastating infectious disease with hundreds of thousands of casualties each year. Antimalarial drug resistance has been a threat to malaria control and elimination for many decades and is still of concern today. Despite the continued effectiveness of current first-line treatments, namely artemisinin-based combination therapies, the emergence of drug-resistant parasites in Southeast Asia and even more alarmingly the occurrence of resistance mutations in Africa is of great concern and requires immediate attention. AREAS COVERED A comprehensive overview of the mechanisms underlying the acquisition of drug resistance in Plasmodium falciparum is given. Understanding these processes provides valuable insights that can be harnessed for the development and selection of novel antimalarials with reduced resistance potential. Additionally, strategies to mitigate resistance to antimalarial compounds on the short term by using approved drugs are discussed. EXPERT OPINION While employing strategies that utilize already approved drugs may offer a prompt and cost-effective approach to counter antimalarial drug resistance, it is crucial to recognize that only continuous efforts into the development of novel antimalarial drugs can ensure the successful treatment of malaria in the future. Incorporating resistance propensity assessment during this developmental process will increase the likelihood of effective and enduring malaria treatments.
Collapse
Affiliation(s)
| | | | - Juliana Inoue
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
| | - Andrea Kreidenweiss
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- German Center for Infection Research (DZIF), Tübingen, Germany
| | - Jana Held
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- German Center for Infection Research (DZIF), Tübingen, Germany
| |
Collapse
|
6
|
Mechanistic basis for multidrug resistance and collateral drug sensitivity conferred to the malaria parasite by polymorphisms in PfMDR1 and PfCRT. PLoS Biol 2022; 20:e3001616. [PMID: 35507548 PMCID: PMC9067703 DOI: 10.1371/journal.pbio.3001616] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 03/31/2022] [Indexed: 01/16/2023] Open
Abstract
Polymorphisms in the Plasmodium falciparum multidrug resistance protein 1 (pfmdr1) gene and the Plasmodium falciparum chloroquine resistance transporter (pfcrt) gene alter the malaria parasite’s susceptibility to most of the current antimalarial drugs. However, the precise mechanisms by which PfMDR1 contributes to multidrug resistance have not yet been fully elucidated, nor is it understood why polymorphisms in pfmdr1 and pfcrt that cause chloroquine resistance simultaneously increase the parasite’s susceptibility to lumefantrine and mefloquine—a phenomenon known as collateral drug sensitivity. Here, we present a robust expression system for PfMDR1 in Xenopus oocytes that enables direct and high-resolution biochemical characterizations of the protein. We show that wild-type PfMDR1 transports diverse pharmacons, including lumefantrine, mefloquine, dihydroartemisinin, piperaquine, amodiaquine, methylene blue, and chloroquine (but not the antiviral drug amantadine). Field-derived mutant isoforms of PfMDR1 differ from the wild-type protein, and each other, in their capacities to transport these drugs, indicating that PfMDR1-induced changes in the distribution of drugs between the parasite’s digestive vacuole (DV) and the cytosol are a key driver of both antimalarial resistance and the variability between multidrug resistance phenotypes. Of note, the PfMDR1 isoforms prevalent in chloroquine-resistant isolates exhibit reduced capacities for chloroquine, lumefantrine, and mefloquine transport. We observe the opposite relationship between chloroquine resistance-conferring mutations in PfCRT and drug transport activity. Using our established assays for characterizing PfCRT in the Xenopus oocyte system and in live parasite assays, we demonstrate that these PfCRT isoforms transport all 3 drugs, whereas wild-type PfCRT does not. We present a mechanistic model for collateral drug sensitivity in which mutant isoforms of PfMDR1 and PfCRT cause chloroquine, lumefantrine, and mefloquine to remain in the cytosol instead of sequestering within the DV. This change in drug distribution increases the access of lumefantrine and mefloquine to their primary targets (thought to be located outside of the DV), while simultaneously decreasing chloroquine’s access to its target within the DV. The mechanistic insights presented here provide a basis for developing approaches that extend the useful life span of antimalarials by exploiting the opposing selection forces they exert upon PfCRT and PfMDR1.
Collapse
|
7
|
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
|
8
|
Wicht KJ, Mok S, Fidock DA. Molecular Mechanisms of Drug Resistance in Plasmodium falciparum Malaria. Annu Rev Microbiol 2021; 74:431-454. [PMID: 32905757 DOI: 10.1146/annurev-micro-020518-115546] [Citation(s) in RCA: 162] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Understanding and controlling the spread of antimalarial resistance, particularly to artemisinin and its partner drugs, is a top priority. Plasmodium falciparum parasites resistant to chloroquine, amodiaquine, or piperaquine harbor mutations in the P. falciparum chloroquine resistance transporter (PfCRT), a transporter resident on the digestive vacuole membrane that in its variant forms can transport these weak-base 4-aminoquinoline drugs out of this acidic organelle, thus preventing these drugs from binding heme and inhibiting its detoxification. The structure of PfCRT, solved by cryogenic electron microscopy, shows mutations surrounding an electronegative central drug-binding cavity where they presumably interact with drugs and natural substrates to control transport. P. falciparum susceptibility to heme-binding antimalarials is also modulated by overexpression or mutations in the digestive vacuole membrane-bound ABC transporter PfMDR1 (P. falciparum multidrug resistance 1 transporter). Artemisinin resistance is primarily mediated by mutations in P. falciparum Kelch13 protein (K13), a protein involved in multiple intracellular processes including endocytosis of hemoglobin, which is required for parasite growth and artemisinin activation. Combating drug-resistant malaria urgently requires the development of new antimalarial drugs with novel modes of action.
Collapse
Affiliation(s)
- Kathryn J Wicht
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, New York 10032, USA; , ,
| | - Sachel Mok
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, New York 10032, USA; , ,
| | - David A Fidock
- 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
| |
Collapse
|
9
|
Expansion of a Specific Plasmodium falciparum PfMDR1 Haplotype in Southeast Asia with Increased Substrate Transport. mBio 2020; 11:mBio.02093-20. [PMID: 33262257 PMCID: PMC7733942 DOI: 10.1128/mbio.02093-20] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Global efforts to eliminate malaria depend on the continued success of artemisinin-based combination therapies (ACTs) that target Plasmodium asexual blood-stage parasites. Resistance to ACTs, however, has emerged, creating the need to define the underlying mechanisms. Mutations in the P. falciparum multidrug resistance protein 1 (PfMDR1) transporter constitute an important determinant of resistance. Applying gene editing tools combined with an analysis of a public database containing thousands of parasite genomes, we show geographic selection and expansion of a pfmdr1 gene amplification encoding the N86/184F haplotype in Southeast Asia. Parasites expressing this PfMDR1 variant possess a higher transport capacity that modulates their responses to antimalarials. These data could help tailor and optimize antimalarial drug usage in different regions where malaria is endemic by taking into account the regional prevalence of pfmdr1 polymorphisms. Artemisinin-based combination therapies (ACTs) have been vital in reducing malaria mortality rates since the 2000s. Their efficacy, however, is threatened by the emergence and spread of artemisinin resistance in Southeast Asia. The Plasmodium falciparum multidrug resistance protein 1 (PfMDR1) transporter plays a central role in parasite resistance to ACT partner drugs through gene copy number variations (CNV) and/or single nucleotide polymorphisms (SNPs). Using genomic epidemiology, we show that multiple pfmdr1 copies encoding the N86 and 184F haplotype are prevalent across Southeast Asia. Applying genome editing tools on the Southeast Asian Dd2 strain and using a surrogate assay to measure transporter activity in infected red blood cells, we demonstrate that parasites harboring multicopy N86/184F PfMDR1 have a higher Fluo-4 transport capacity compared with those expressing the wild-type N86/Y184 haplotype. Multicopy N86/184F PfMDR1 is also associated with decreased parasite susceptibility to lumefantrine. These findings provide evidence of the geographic selection and expansion of specific multicopy PfMDR1 haplotypes associated with multidrug resistance in Southeast Asia.
Collapse
|
10
|
Shibeshi MA, Kifle ZD, Atnafie SA. Antimalarial Drug Resistance and Novel Targets for Antimalarial Drug Discovery. Infect Drug Resist 2020; 13:4047-4060. [PMID: 33204122 PMCID: PMC7666977 DOI: 10.2147/idr.s279433] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 10/29/2020] [Indexed: 12/16/2022] Open
Abstract
Malaria is among the most devastating and widespread tropical parasitic diseases in which most prevalent in developing countries. Antimalarial drug resistance is the ability of a parasite strain to survive and/or to multiply despite the administration and absorption of medicine given in doses equal to or higher than those usually recommended. Among the factors which facilitate the emergence of resistance to existing antimalarial drugs: the parasite mutation rate, the overall parasite load, the strength of drug selected, the treatment compliance, poor adherence to malaria treatment guideline, improper dosing, poor pharmacokinetic properties, fake drugs lead to inadequate drug exposure on parasites, and poor-quality antimalarial may aid and abet resistance. Malaria vaccines can be categorized into three categories: pre-erythrocytic, blood-stage, and transmission-blocking vaccines. Molecular markers of antimalarial drug resistance are used to screen for the emergence of resistance and assess its spread. It provides information about the parasite genetics associated with resistance, either single nucleotide polymorphisms or gene copy number variations which are associated with decreased susceptibility of parasites to antimalarial drugs. Glucose transporter PfHT1, kinases (Plasmodium kinome), food vacuole, apicoplast, cysteine proteases, and aminopeptidases are the novel targets for the development of new antimalarial drugs. Therefore, this review summarizes the antimalarial drug resistance and novel targets of antimalarial drugs.
Collapse
Affiliation(s)
- Melkamu Adigo Shibeshi
- Department of Pharmacology, School of Pharmacy, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Zemene Demelash Kifle
- Department of Pharmacology, School of Pharmacy, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Seyfe Asrade Atnafie
- Department of Pharmacology, School of Pharmacy, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
11
|
Mata-Cantero L, Chaparro MJ, Colmenarejo G, Cid C, Cortes Cabrera A, Rodriguez MS, Martín J, Gamo FJ, Gomez-Lorenzo MG. Identification of Small Molecules Disrupting the Ubiquitin Proteasome System in Malaria. ACS Infect Dis 2019; 5:2105-2117. [PMID: 31644867 DOI: 10.1021/acsinfecdis.9b00216] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The ubiquitin proteasome system (UPS) is one of the main proteolytic pathways in eukaryotic cells, playing an essential role in key cellular processes such as cell cycling and signal transduction. Changes in some of the components of this pathway have been implicated in various conditions, including cancer and infectious diseases such as malaria. The success of therapies based on proteasome inhibitors has been shown in human clinical trials. In addition to its proven tractability, the essentiality of the Plasmodium falciparum UPS underlines its potential as a source of targets to identify new antimalarial treatments. Two assays, previously developed to quantify the parasite protein ubiquitylation levels in a high throughput format, have been used to identify compounds that inhibit parasite growth by targeting P. falciparum UPS. Among the positive hits, specific inhibitors of the P. falciparum proteasome have been identified and characterized. Hits identified using this approach may be used as starting points for development of new antimalarial drugs. They may also be used as tools to further understand proteasome function and to identify new targets in P. falciparum UPS.
Collapse
Affiliation(s)
- Lydia Mata-Cantero
- Tres Cantos Medicines Development Campus, Diseases of the Developing World. GlaxoSmithKline, Severo Ochoa 2, Tres Cantos, 28760 Madrid, Spain
| | - María Jesús Chaparro
- Tres Cantos Medicines Development Campus, Diseases of the Developing World. GlaxoSmithKline, Severo Ochoa 2, Tres Cantos, 28760 Madrid, Spain
| | - Gonzalo Colmenarejo
- Tres Cantos Medicines Development Campus, Diseases of the Developing World. GlaxoSmithKline, Severo Ochoa 2, Tres Cantos, 28760 Madrid, Spain
- IMDEA Food, Biostatistics and Bioinformatics Unit, Ctra Cantoblanco 8, 28049 Madrid, Spain
| | - Concepción Cid
- Tres Cantos Medicines Development Campus, Diseases of the Developing World. GlaxoSmithKline, Severo Ochoa 2, Tres Cantos, 28760 Madrid, Spain
| | - Alvaro Cortes Cabrera
- Tres Cantos Medicines Development Campus, Diseases of the Developing World. GlaxoSmithKline, Severo Ochoa 2, Tres Cantos, 28760 Madrid, Spain
| | - Manuel S. Rodriguez
- Université de Toulouse, ITAV CNRS and IPBS CNRS, 1 place Pierre Potier, Oncopole entrée B, 31106 Toulouse, France
| | - Julio Martín
- Tres Cantos Medicines Development Campus, Diseases of the Developing World. GlaxoSmithKline, Severo Ochoa 2, Tres Cantos, 28760 Madrid, Spain
| | - Francisco Javier Gamo
- Tres Cantos Medicines Development Campus, Diseases of the Developing World. GlaxoSmithKline, Severo Ochoa 2, Tres Cantos, 28760 Madrid, Spain
| | - Maria G. Gomez-Lorenzo
- Tres Cantos Medicines Development Campus, Diseases of the Developing World. GlaxoSmithKline, Severo Ochoa 2, Tres Cantos, 28760 Madrid, Spain
| |
Collapse
|
12
|
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
|
13
|
Cowell AN, Winzeler EA. The genomic architecture of antimalarial drug resistance. Brief Funct Genomics 2019; 18:314-328. [PMID: 31119263 PMCID: PMC6859814 DOI: 10.1093/bfgp/elz008] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 03/19/2019] [Accepted: 04/09/2019] [Indexed: 11/15/2022] Open
Abstract
Plasmodium falciparum and Plasmodium vivax, the two protozoan parasite species that cause the majority of cases of human malaria, have developed resistance to nearly all known antimalarials. The ability of malaria parasites to develop resistance is primarily due to the high numbers of parasites in the infected person's bloodstream during the asexual blood stage of infection in conjunction with the mutability of their genomes. Identifying the genetic mutations that mediate antimalarial resistance has deepened our understanding of how the parasites evade our treatments and reveals molecular markers that can be used to track the emergence of resistance in clinical samples. In this review, we examine known genetic mutations that lead to resistance to the major classes of antimalarial medications: the 4-aminoquinolines (chloroquine, amodiaquine and piperaquine), antifolate drugs, aryl amino-alcohols (quinine, lumefantrine and mefloquine), artemisinin compounds, antibiotics (clindamycin and doxycycline) and a napthoquinone (atovaquone). We discuss how the evolution of antimalarial resistance informs strategies to design the next generation of antimalarial therapies.
Collapse
Affiliation(s)
- Annie N Cowell
- Division of Infectious Diseases and Global Health, Department of Medicine, University of California, San Diego, Gilman Dr., La Jolla, CA, USA
| | - Elizabeth A Winzeler
- Division of Host-Microbe Systems & Therapeutics, Department of Pediatrics, University of California, San Diego, Gilman Dr., La Jolla, CA, USA
| |
Collapse
|
14
|
Idowu AO, Oyibo WA, Bhattacharyya S, Khubbar M, Mendie UE, Bumah VV, Black C, Igietseme J, Azenabor AA. Rare mutations in Pfmdr1 gene of Plasmodium falciparum detected in clinical isolates from patients treated with anti-malarial drug in Nigeria. Malar J 2019; 18:319. [PMID: 31533729 PMCID: PMC6751857 DOI: 10.1186/s12936-019-2947-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 09/06/2019] [Indexed: 01/18/2023] Open
Abstract
Background Plasmodium falciparum, the deadliest causative agent of malaria, has high prevalence in Nigeria. Drug resistance causing failure of previously effective drugs has compromised anti-malarial treatment. On this basis, there is need for a proactive surveillance for resistance markers to the currently recommended artemisinin-based combination therapy (ACT), for early detection of resistance before it become widespread. Methods This study assessed anti-malarial resistance genes polymorphism in patients with uncomplicated P. falciparum malaria in Lagos, Nigeria. Sanger and Next Generation Sequencing (NGS) methods were used to screen for mutations in thirty-seven malaria positive blood samples targeting the P. falciparum chloroquine-resistance transporter (Pfcrt), P. falciparum multidrug-resistance 1 (Pfmdr1), and P. falciparum kelch 13 (Pfk13) genes, which have been previously associated with anti-malarial resistance. Results Expectedly, the NGS method was more proficient, detecting six Pfmdr1, seven Pfcrt and three Pfk13 mutations in the studied clinical isolates from Nigeria, a malaria endemic area. These mutations included rare Pfmdr1 mutations, N504K, N649D, F938Y and S967N, which were previously unreported. In addition, there was moderate prevalence of the K76T mutation (34.6%) associated with chloroquine and amodiaquine resistance, and high prevalence of the N86 wild type allele (92.3%) associated with lumefantrine resistance. Conclusion Widespread circulation of mutations associated with resistance to current anti-malarial drugs could potentially limit effective malaria therapy in endemic populations.
Collapse
Affiliation(s)
- Abel O Idowu
- Department of Biomedical Sciences, College of Health Sciences, University of Wisconsin, 2400 E. Hartford Avenue, Milwaukee, WI, 53211, USA.,Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, University of Lagos, Lagos, Nigeria
| | - Wellington A Oyibo
- ANDI Centre of Excellence in Malaria Diagnosis, College of Medicine, University of Lagos, Lagos, Nigeria
| | | | - Manjeet Khubbar
- City of Milwaukee Health Department Laboratory, Milwaukee, USA
| | - Udoma E Mendie
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, University of Lagos, Lagos, Nigeria
| | - Violet V Bumah
- Department of Biology, North Life Science 317, San Diego State University, San Diego, CA, 92182, USA
| | - Carolyn Black
- Molecular Pathogenesis Laboratory, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Joseph Igietseme
- Molecular Pathogenesis Laboratory, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Anthony A Azenabor
- Department of Biomedical Sciences, College of Health Sciences, University of Wisconsin, 2400 E. Hartford Avenue, Milwaukee, WI, 53211, USA. .,Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, University of Lagos, Lagos, Nigeria.
| |
Collapse
|
15
|
Wong W, Bai XC, Sleebs BE, Triglia T, Brown A, Thompson JK, Jackson KE, Hanssen E, Marapana DS, Fernandez IS, Ralph SA, Cowman AF, Scheres SH, Baum J. Mefloquine targets the Plasmodium falciparum 80S ribosome to inhibit protein synthesis. Nat Microbiol 2017; 2:17031. [PMID: 28288098 PMCID: PMC5439513 DOI: 10.1038/nmicrobiol.2017.31] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 02/07/2017] [Indexed: 12/26/2022]
Abstract
Malaria control is heavily dependent on chemotherapeutic agents for disease prevention and drug treatment. Defining the mechanism of action for licensed drugs, for which no target is characterized, is critical to the development of their second-generation derivatives to improve drug potency towards inhibition of their molecular targets. Mefloquine is a widely used antimalarial without a known mode of action. Here, we demonstrate that mefloquine is a protein synthesis inhibitor. We solved a 3.2 Å cryo-electron microscopy structure of the Plasmodium falciparum 80S ribosome with the (+)-mefloquine enantiomer bound to the ribosome GTPase-associated centre. Mutagenesis of mefloquine-binding residues generates parasites with increased resistance, confirming the parasite-killing mechanism. Furthermore, structure-guided derivatives with an altered piperidine group, predicted to improve binding, show enhanced parasiticidal effect. These data reveal one possible mode of action for mefloquine and demonstrate the vast potential of cryo-electron microscopy to guide the development of mefloquine derivatives to inhibit parasite protein synthesis.
Collapse
Affiliation(s)
- Wilson Wong
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia
| | - Xiao-Chen Bai
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge CB2 0QH, UK
| | - Brad E. Sleebs
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia
| | - Tony Triglia
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia
| | - Alan Brown
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge CB2 0QH, UK
| | - Jennifer K. Thompson
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia
| | - Katherine E. Jackson
- Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Eric Hanssen
- Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Danushka S. Marapana
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia
| | - Israel S. Fernandez
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge CB2 0QH, UK
| | - Stuart A. Ralph
- Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Alan F. Cowman
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Sjors H.W. Scheres
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge CB2 0QH, UK
| | - Jake Baum
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, 3010, Australia
- Department of Life Sciences, Imperial College London, South Kensington, London, SW7 2AZ, UK
| |
Collapse
|
16
|
Parhizgar AR, Tahghighi A. Introducing New Antimalarial Analogues of Chloroquine and Amodiaquine: A Narrative Review. IRANIAN JOURNAL OF MEDICAL SCIENCES 2017; 42:115-128. [PMID: 28360437 PMCID: PMC5366359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 05/02/2016] [Accepted: 06/05/2016] [Indexed: 11/03/2022]
Abstract
Antimalarial drugs with the 4-aminoquinoline scaffold such as the important drugs, chloroquine (CQ) and amodiaquine (AQ), have been used to prevent and treat malaria for many years. The importance of these drugs is related to their simple usage, high efficacy, affordability, and cost-effectiveness of their synthesis. In recent years, with the spread of parasite resistance to CQ and cross-resistance to its other analogues have decreased their consumption in many geographical areas. On the other hand, AQ is an effective antimalarial drug which its usage has been restricted due to hepatic and hematological toxicities. The significance of the quinoline ring at quinoline-based antimalarial drugs has prompted research centers and pharmaceutical companies to focus on the design and synthesis of new analogues of these drugs, especially CQ and AQ analogues. Accordingly, various derivatives have been synthesized and evaluated in vitro and in vivo against the resistant strains of the malaria parasite to solve the problem of drug resistance. Also, the pharmacokinetic properties of these compounds have been evaluated to augment their efficacy and diminish their toxicity. Some of these analogues are currently in clinical and preclinical development. Consequently, the recent researches showed yet 4-aminoquinoline scaffold is active moiety in new compounds with antiplasmodial activity. Hence, the aim of this review article is to introduce of the novel synthetic analogues of CQ and AQ, which may constitute the next generation of antimalarial drugs with the 4-aminoquinoline scaffold.
Collapse
Affiliation(s)
- Arezoo Rafiee Parhizgar
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
- Department of Medicinal Chemistry, Pharmaceutical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Azar Tahghighi
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
17
|
Witkowski B, Duru V, Khim N, Ross LS, Saintpierre B, Beghain J, Chy S, Kim S, Ke S, Kloeung N, Eam R, Khean C, Ken M, Loch K, Bouillon A, Domergue A, Ma L, Bouchier C, Leang R, Huy R, Nuel G, Barale JC, Legrand E, Ringwald P, Fidock DA, Mercereau-Puijalon O, Ariey F, Ménard D. A surrogate marker of piperaquine-resistant Plasmodium falciparum malaria: a phenotype-genotype association study. THE LANCET. INFECTIOUS DISEASES 2016; 17:174-183. [PMID: 27818097 PMCID: PMC5266792 DOI: 10.1016/s1473-3099(16)30415-7] [Citation(s) in RCA: 269] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 09/26/2016] [Accepted: 09/30/2016] [Indexed: 11/30/2022]
Abstract
Background Western Cambodia is the epicentre of Plasmodium falciparum multidrug resistance and is facing high rates of dihydroartemisinin–piperaquine treatment failures. Genetic tools to detect the multidrug-resistant parasites are needed. Artemisinin resistance can be tracked using the K13 molecular marker, but no marker exists for piperaquine resistance. We aimed to identify genetic markers of piperaquine resistance and study their association with dihydroartemisinin–piperaquine treatment failures. Methods We obtained blood samples from Cambodian patients infected with P falciparum and treated with dihydroartemisinin–piperaquine. Patients were followed up for 42 days during the years 2009–15. We established in-vitro and ex-vivo susceptibility profiles for a subset using piperaquine survival assays. We determined whole-genome sequences by Illumina paired-reads sequencing, copy number variations by qPCR, RNA concentrations by qRT-PCR, and protein concentrations by immunoblotting. Fisher’s exact and non-parametric Wilcoxon rank-sum tests were used to identify significant differences in single-nucleotide polymorphisms or copy number variants, respectively, for differential distribution between piperaquine-resistant and piperaquine-sensitive parasite lines. Findings Whole-genome exon sequence analysis of 31 culture-adapted parasite lines associated amplification of the plasmepsin 2–plasmepsin 3 gene cluster with in-vitro piperaquine resistance. Ex-vivo piperaquine survival assay profiles of 134 isolates correlated with plasmepsin 2 gene copy number. In 725 patients treated with dihydroartemisinin–piperaquine, multicopy plasmepsin 2 in the sample collected before treatment was associated with an adjusted hazard ratio (aHR) for treatment failure of 20·4 (95% CI 9·1–45·5, p<0·0001). Multicopy plasmepsin 2 predicted dihydroartemisinin–piperaquine failures with 0·94 (95% CI 0·88–0·98) sensitivity and 0·77 (0·74–0·81) specificity. Analysis of samples collected across the country from 2002 to 2015 showed that the geographical and temporal increase of the proportion of multicopy plasmepsin 2 parasites was highly correlated with increasing dihydroartemisinin–piperaquine treatment failure rates (r=0·89 [95% CI 0·77–0·95], p<0·0001, Spearman’s coefficient of rank correlation). Dihydroartemisinin–piperaquine efficacy at day 42 fell below 90% when the proportion of multicopy plasmepsin 2 parasites exceeded 22%. Interpretation Piperaquine resistance in Cambodia is strongly associated with amplification of plasmepsin 2–3, encoding haemoglobin-digesting proteases, regardless of the location. Multicopy plasmepsin 2 constitutes a surrogate molecular marker to track piperaquine resistance. A molecular toolkit combining plasmepsin 2 with K13 and mdr1 monitoring should provide timely information for antimalarial treatment and containment policies. Funding Institut Pasteur in Cambodia, Institut Pasteur Paris, National Institutes of Health, WHO, Agence Nationale de la Recherche, Investissement d’Avenir programme, Laboratoire d’Excellence Integrative “Biology of Emerging Infectious Diseases”.
Collapse
Affiliation(s)
- Benoit Witkowski
- Malaria Molecular Epidemiology Unit, Institut Pasteur in Cambodia, Phnom Penh, Cambodia; Malaria Translational Research Unit, Institut Pasteur, Paris, France; Institut Pasteur in Cambodia, Phnom Penh, Cambodia
| | - Valentine Duru
- Malaria Molecular Epidemiology Unit, Institut Pasteur in Cambodia, Phnom Penh, Cambodia
| | - Nimol Khim
- Malaria Molecular Epidemiology Unit, Institut Pasteur in Cambodia, Phnom Penh, Cambodia; Malaria Translational Research Unit, Institut Pasteur, Paris, France; Institut Pasteur in Cambodia, Phnom Penh, Cambodia
| | - Leila S Ross
- Department of Microbiology and Immunology and Division of Infectious Diseases, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | | | - Johann Beghain
- Department of Parasites and Insect Vectors, Institut Pasteur, Paris, France
| | - Sophy Chy
- Malaria Molecular Epidemiology Unit, Institut Pasteur in Cambodia, Phnom Penh, Cambodia
| | - Saorin Kim
- Malaria Molecular Epidemiology Unit, Institut Pasteur in Cambodia, Phnom Penh, Cambodia
| | - Sopheakvatey Ke
- Malaria Molecular Epidemiology Unit, Institut Pasteur in Cambodia, Phnom Penh, Cambodia
| | - Nimol Kloeung
- Malaria Molecular Epidemiology Unit, Institut Pasteur in Cambodia, Phnom Penh, Cambodia
| | - Rotha Eam
- Malaria Molecular Epidemiology Unit, Institut Pasteur in Cambodia, Phnom Penh, Cambodia
| | - Chanra Khean
- Malaria Molecular Epidemiology Unit, Institut Pasteur in Cambodia, Phnom Penh, Cambodia
| | - Malen Ken
- Malaria Molecular Epidemiology Unit, Institut Pasteur in Cambodia, Phnom Penh, Cambodia
| | - Kaknika Loch
- Malaria Molecular Epidemiology Unit, Institut Pasteur in Cambodia, Phnom Penh, Cambodia
| | - Anthony Bouillon
- Malaria Translational Research Unit, Institut Pasteur, Paris, France; Institut Pasteur in Cambodia, Phnom Penh, Cambodia; Structural Microbiology Unit, Biology of Malaria Targets Group, Department of Structural Biology and Chemistry and CNRS, UMR3528, Institut Pasteur, Paris, France
| | - Anais Domergue
- Malaria Molecular Epidemiology Unit, Institut Pasteur in Cambodia, Phnom Penh, Cambodia
| | - Laurence Ma
- Plate-forme Génomique, Département Génomes et Génétique, Institut Pasteur, Paris, France
| | - Christiane Bouchier
- Plate-forme Génomique, Département Génomes et Génétique, Institut Pasteur, Paris, France
| | - Rithea Leang
- National Center for Parasitology, Entomology and Malaria Control, Phnom Penh, Cambodia
| | - Rekol Huy
- National Center for Parasitology, Entomology and Malaria Control, Phnom Penh, Cambodia
| | - Grégory Nuel
- Laboratoire de Mathématiques Appliquées (MAP5) UMR CNRS 8145, Université Paris Descartes, Paris, France
| | - Jean-Christophe Barale
- Malaria Translational Research Unit, Institut Pasteur, Paris, France; Institut Pasteur in Cambodia, Phnom Penh, Cambodia; Structural Microbiology Unit, Biology of Malaria Targets Group, Department of Structural Biology and Chemistry and CNRS, UMR3528, Institut Pasteur, Paris, France
| | - Eric Legrand
- Malaria Translational Research Unit, Institut Pasteur, Paris, France; Institut Pasteur in Cambodia, Phnom Penh, Cambodia; Department of Parasites and Insect Vectors, Institut Pasteur, Paris, France
| | - Pascal Ringwald
- Global Malaria Programme, World Health Organization, Geneva, Switzerland
| | - David A Fidock
- Department of Microbiology and Immunology and Division of Infectious Diseases, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | | | - Frédéric Ariey
- Department of Parasites and Insect Vectors, Institut Pasteur, Paris, France; Institut Cochin Inserm U1016, Université Paris-Descartes, Sorbonne Paris Cité, and Laboratoire de Parasitologie-Mycologie, Hôpital Cochin, Paris, France
| | - Didier Ménard
- Malaria Molecular Epidemiology Unit, Institut Pasteur in Cambodia, Phnom Penh, Cambodia; Malaria Translational Research Unit, Institut Pasteur, Paris, France; Institut Pasteur in Cambodia, Phnom Penh, Cambodia.
| |
Collapse
|
18
|
Xu C, Wei QK, Li J, Xiao T, Yin K, Zhao CL, Wang YB, Kong XL, Zhao GH, Sun H, Liu X, Huang BC. Characteristics of Imported Malaria and Species of Plasmodium Involved in Shandong Province, China (2012-2014). THE KOREAN JOURNAL OF PARASITOLOGY 2016; 54:407-14. [PMID: 27658591 PMCID: PMC5040091 DOI: 10.3347/kjp.2016.54.4.407] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 05/19/2016] [Accepted: 05/24/2016] [Indexed: 02/04/2023]
Abstract
Malaria remains a serious public health problem in Shandong Province, China; therefore, it is important to explore the characteristics of the current malaria prevalence situation in the province. In this study, data of malaria cases reported in Shandong during 2012-2014 were analyzed, and Plasmodium species were confirmed by smear microscopy and nested-PCR. A total of 374 malaria cases were reported, 80.8% of which were reported from 6 prefectures. Of all cases, P. falciparum was dominant (81.3%), followed by P. vivax (11.8%); P. ovale and P. malariae together accounted for 6.4% of cases. Notably, for the first time since 2012, no indigenous case had been reported in Shandong Province, a situation that continued through 2014. Total 95.2% of cases were imported from Africa. The ratio of male/female was 92.5:1, and 96.8% of cases occurred in people 20-54 years of age. Farmers or laborers represented 77.5% of cases. No significant trends of monthly pattern were found in the reported cases. All patients were in good condition after treatment, except for 3 who died. These results indicate that imported malaria has increased significantly since 2012 in Shandong Province, especially for P. falciparum, and there is an emergence of species diversity.
Collapse
Affiliation(s)
- Chao Xu
- Shandong Academy of Medical Sciences, Shandong Institute of Parasitic Diseases, Shandong Provincial Reference Laboratory for Malaria Diagnosis, Jining, Shandong Province 272033, People's Republic of China
| | - Qing-Kuan Wei
- Shandong Academy of Medical Sciences, Shandong Institute of Parasitic Diseases, Shandong Provincial Reference Laboratory for Malaria Diagnosis, Jining, Shandong Province 272033, People's Republic of China
| | - Jin Li
- Shandong Academy of Medical Sciences, Shandong Institute of Parasitic Diseases, Shandong Provincial Reference Laboratory for Malaria Diagnosis, Jining, Shandong Province 272033, People's Republic of China
| | - Ting Xiao
- Shandong Academy of Medical Sciences, Shandong Institute of Parasitic Diseases, Shandong Provincial Reference Laboratory for Malaria Diagnosis, Jining, Shandong Province 272033, People's Republic of China
| | - Kun Yin
- Shandong Academy of Medical Sciences, Shandong Institute of Parasitic Diseases, Shandong Provincial Reference Laboratory for Malaria Diagnosis, Jining, Shandong Province 272033, People's Republic of China
| | - Chang-Lei Zhao
- Shandong Academy of Medical Sciences, Shandong Institute of Parasitic Diseases, Shandong Provincial Reference Laboratory for Malaria Diagnosis, Jining, Shandong Province 272033, People's Republic of China
| | - Yong-Bin Wang
- Shandong Academy of Medical Sciences, Shandong Institute of Parasitic Diseases, Shandong Provincial Reference Laboratory for Malaria Diagnosis, Jining, Shandong Province 272033, People's Republic of China
| | - Xiang-Li Kong
- Shandong Academy of Medical Sciences, Shandong Institute of Parasitic Diseases, Shandong Provincial Reference Laboratory for Malaria Diagnosis, Jining, Shandong Province 272033, People's Republic of China
| | - Gui-Hua Zhao
- Shandong Academy of Medical Sciences, Shandong Institute of Parasitic Diseases, Shandong Provincial Reference Laboratory for Malaria Diagnosis, Jining, Shandong Province 272033, People's Republic of China
| | - Hui Sun
- Shandong Academy of Medical Sciences, Shandong Institute of Parasitic Diseases, Shandong Provincial Reference Laboratory for Malaria Diagnosis, Jining, Shandong Province 272033, People's Republic of China
| | - Xin Liu
- Shandong Academy of Medical Sciences, Shandong Institute of Parasitic Diseases, Shandong Provincial Reference Laboratory for Malaria Diagnosis, Jining, Shandong Province 272033, People's Republic of China
| | - Bing-Cheng Huang
- Shandong Academy of Medical Sciences, Shandong Institute of Parasitic Diseases, Shandong Provincial Reference Laboratory for Malaria Diagnosis, Jining, Shandong Province 272033, People's Republic of China
| |
Collapse
|
19
|
Globally prevalent PfMDR1 mutations modulate Plasmodium falciparum susceptibility to artemisinin-based combination therapies. Nat Commun 2016; 7:11553. [PMID: 27189525 PMCID: PMC4873939 DOI: 10.1038/ncomms11553] [Citation(s) in RCA: 205] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 04/07/2016] [Indexed: 02/07/2023] Open
Abstract
Antimalarial chemotherapy, globally reliant on artemisinin-based combination therapies (ACTs), is threatened by the spread of drug resistance in Plasmodium falciparum parasites. Here we use zinc-finger nucleases to genetically modify the multidrug resistance-1 transporter PfMDR1 at amino acids 86 and 184, and demonstrate that the widely prevalent N86Y mutation augments resistance to the ACT partner drug amodiaquine and the former first-line agent chloroquine. In contrast, N86Y increases parasite susceptibility to the partner drugs lumefantrine and mefloquine, and the active artemisinin metabolite dihydroartemisinin. The PfMDR1 N86 plus Y184F isoform moderately reduces piperaquine potency in strains expressing an Asian/African variant of the chloroquine resistance transporter PfCRT. Mutations in both digestive vacuole-resident transporters are thought to differentially regulate ACT drug interactions with host haem, a product of parasite-mediated haemoglobin degradation. Global mapping of these mutations illustrates where the different ACTs could be selectively deployed to optimize treatment based on regional differences in PfMDR1 haplotypes. Antimalarial chemotherapy relies on combination therapies (ACTs) consisting of an artemisinin derivative and a partner drug. Here, the authors study the effects of globally prevalent mutations in a multidrug resistance transporter (PfMDR1) on the parasite's susceptibility to ACT drugs.
Collapse
|
20
|
Ng CL, Siciliano G, Lee MCS, de Almeida MJ, Corey VC, Bopp SE, Bertuccini L, Wittlin S, Kasdin RG, Le Bihan A, Clozel M, Winzeler EA, Alano P, Fidock DA. CRISPR-Cas9-modified pfmdr1 protects Plasmodium falciparum asexual blood stages and gametocytes against a class of piperazine-containing compounds but potentiates artemisinin-based combination therapy partner drugs. Mol Microbiol 2016; 101:381-93. [PMID: 27073104 DOI: 10.1111/mmi.13397] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/10/2016] [Indexed: 01/08/2023]
Abstract
Emerging resistance to first-line antimalarial combination therapies threatens malaria treatment and the global elimination campaign. Improved therapeutic strategies are required to protect existing drugs and enhance treatment efficacy. We report that the piperazine-containing compound ACT-451840 exhibits single-digit nanomolar inhibition of the Plasmodium falciparum asexual blood stages and transmissible gametocyte forms. Genome sequence analyses of in vitro-derived ACT-451840-resistant parasites revealed single nucleotide polymorphisms in pfmdr1, which encodes a digestive vacuole membrane-bound ATP-binding cassette transporter known to alter P. falciparum susceptibility to multiple first-line antimalarials. CRISPR-Cas9 based gene editing confirmed that PfMDR1 point mutations mediated ACT-451840 resistance. Resistant parasites demonstrated increased susceptibility to the clinical drugs lumefantrine, mefloquine, quinine and amodiaquine. Stage V gametocytes harboring Cas9-introduced pfmdr1 mutations also acquired ACT-451840 resistance. These findings reveal that PfMDR1 mutations can impart resistance to compounds active against asexual blood stages and mature gametocytes. Exploiting PfMDR1 resistance mechanisms provides new opportunities for developing disease-relieving and transmission-blocking antimalarials.
Collapse
Affiliation(s)
- Caroline L Ng
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Giulia Siciliano
- Dipartimento di Malattie Infettive, Parassitarie ed Immunomediate, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Marcus C S Lee
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Mariana J de Almeida
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Victoria C Corey
- Division of Host-Microbe Systems & Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, CA, 92093, USA
| | - Selina E Bopp
- Division of Host-Microbe Systems & Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, CA, 92093, USA
| | - Lucia Bertuccini
- Dipartimento Tecnologie e Salute, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Sergio Wittlin
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, CH-4002 Basel, Switzerland
| | - Rachel G Kasdin
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Amélie Le Bihan
- Department of Drug Discovery, Actelion Pharmaceuticals Ltd., CH-4123 Allschwil, Switzerland
| | - Martine Clozel
- Department of Drug Discovery, Actelion Pharmaceuticals Ltd., CH-4123 Allschwil, Switzerland
| | - Elizabeth A Winzeler
- Division of Host-Microbe Systems & Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, CA, 92093, USA
| | - Pietro Alano
- Dipartimento di Malattie Infettive, Parassitarie ed Immunomediate, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - David A Fidock
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY, 10032, USA.,Division of Infectious Diseases, Department of Medicine, Columbia University Medical Center, New York, NY, 10032, USA
| |
Collapse
|
21
|
Spillman NJ, Kirk K. The malaria parasite cation ATPase PfATP4 and its role in the mechanism of action of a new arsenal of antimalarial drugs. INTERNATIONAL JOURNAL FOR PARASITOLOGY-DRUGS AND DRUG RESISTANCE 2015; 5:149-62. [PMID: 26401486 PMCID: PMC4559606 DOI: 10.1016/j.ijpddr.2015.07.001] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 07/03/2015] [Accepted: 07/08/2015] [Indexed: 11/28/2022]
Abstract
The intraerythrocytic malaria parasite, Plasmodium falciparum, maintains a low cytosolic Na(+) concentration and the plasma membrane P-type cation translocating ATPase 'PfATP4' has been implicated as playing a key role in this process. PfATP4 has been the subject of significant attention in recent years as mutations in this protein confer resistance to a growing number of new antimalarial compounds, including the spiroindolones, the pyrazoles, the dihydroisoquinolones, and a number of the antimalarial agents in the Medicines for Malaria Venture's 'Malaria Box'. On exposure of parasites to these compounds there is a rapid disruption of cytosolic Na(+). Whether, and if so how, such chemically distinct compounds interact with PfATP4, and how such interactions lead to parasite death, is not yet clear. The fact that multiple different chemical classes have converged upon PfATP4 highlights its significance as a potential target for new generation antimalarial agents. A spiroindolone (KAE609, now known as cipargamin) has progressed through Phase I and IIa clinical trials with favourable results. In this review we consider the physiological role of PfATP4, summarise the current repertoire of antimalarial compounds for which PfATP4 is implicated in their mechanism of action, and provide an outlook on translation from target identification in the laboratory to patient treatment in the field.
Collapse
Affiliation(s)
- Natalie Jane Spillman
- Research School of Biology, The Australian National University, Canberra, Australian Capital Territory 2601, Australia ; Department of Medicine (Infectious Diseases), Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Kiaran Kirk
- Research School of Biology, The Australian National University, Canberra, Australian Capital Territory 2601, Australia
| |
Collapse
|
22
|
Reiling SJ, Rohrbach P. Monitoring PfMDR1 transport in Plasmodium falciparum. Malar J 2015; 14:270. [PMID: 26169590 PMCID: PMC4501111 DOI: 10.1186/s12936-015-0791-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 07/01/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The Plasmodium falciparum multidrug resistance 1 transporter, PfMDR1, contains five amino acid polymorphisms that are suggested to be involved in altered drug transport from the parasite's cytosol into the digestive vacuole (DV). Transport of a substrate into another intracellular compartment influences drug availability at its site of action, therefore making the parasite more susceptible or resistant to a drug. Fluo-4 is a known fluorescent substrate that can be used as a molecular tool to investigate transport dynamics of PfMDR1 in many parasite strains. METHODS Six P. falciparum strains with varying PfMDR1 mutations were loaded with Fluo-4 AM. Accumulation of the fluorophore in the DV was measured using confocal microscopy. The role of a key amino acid mutation was verified using selected parasite clones with point mutations at PfMDR1 amino acid position 1042. Equal expression of PfMDR1 was confirmed by Western blot. RESULTS Fluo-4 was transported by PfMDR1 into the DV of most drug-sensitive and -resistant parasites. Asparagine at PfMDR1 amino acid position 1042 was crucial for Fluo-4 transport, while the N1042D substitution abolished Fluo-4 transport. Competition studies of Fluo-4 with chloroquine, quinine and mefloquine were performed on parasites harbouring asparagine at position 1042. A distinct Fluo-4 transport inhibition pattern for each tested anti-malarial drug was observed in parasite strains of different genetic background. CONCLUSION This study demonstrates that Fluo-4 can be used to investigate PfMDR1 transport dynamics in both drug-sensitive and -resistant parasites. Furthermore, direct evidence of altered Fluo-4 transport in PfMDR1 is linked to a single amino acid mutation in the substrate binding pocket. This system offers a great tool to investigate the role of substrate transport by PfMDR1 and the mutations necessary to support transport, which would lead to new insights for the development of novel anti-malarial drugs.
Collapse
Affiliation(s)
- Sarah J Reiling
- Institute of Parasitology, McGill University, Ste. Anne de Bellevue (Montreal), QC, H9X-3V9, Canada.
| | - Petra Rohrbach
- Institute of Parasitology, McGill University, Ste. Anne de Bellevue (Montreal), QC, H9X-3V9, Canada.
| |
Collapse
|
23
|
Role of Different Pfcrt and Pfmdr-1 Mutations in Conferring Resistance to Antimalaria Drugs in Plasmodium falciparum. Malar Res Treat 2014; 2014:950424. [PMID: 25506039 PMCID: PMC4243603 DOI: 10.1155/2014/950424] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Accepted: 08/30/2014] [Indexed: 01/28/2023] Open
Abstract
Emergence of drugs resistant strains of Plasmodium falciparum has augmented the scourge of malaria in endemic areas. Antimalaria drugs act on different intracellular targets. The majority of them interfere with digestive vacuoles (DVs) while others affect other organelles, namely, apicoplast and mitochondria. Prevention of drug accumulation or access into the target site is one of the mechanisms that plasmodium adopts to develop resistance. Plasmodia are endowed with series of transporters that shuffle drugs away from the target site, namely, pfmdr (Plasmodium falciparum multidrug resistance transporter) and pfcrt (Plasmodium falciparum chloroquine resistance transporter) which exist in DV membrane and are considered as putative markers of CQ resistance. They are homologues to human P-glycoproteins (P-gh or multidrug resistance system) and members of drug metabolite transporter (DMT) family, respectively. The former mediates drifting of xenobiotics towards the DV while the latter chucks them outside. Resistance to drugs whose target site of action is intravacuolar develops when the transporters expel them outside the DVs and vice versa for those whose target is extravacuolar. In this review, we are going to summarize the possible pfcrt and pfmdr mutation and their role in changing plasmodium sensitivity to different anti-Plasmodium drugs.
Collapse
|
24
|
Sanchez CP, Liu CH, Mayer S, Nurhasanah A, Cyrklaff M, Mu J, Ferdig MT, Stein WD, Lanzer M. A HECT ubiquitin-protein ligase as a novel candidate gene for altered quinine and quinidine responses in Plasmodium falciparum. PLoS Genet 2014; 10:e1004382. [PMID: 24830312 PMCID: PMC4022464 DOI: 10.1371/journal.pgen.1004382] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 04/01/2014] [Indexed: 11/18/2022] Open
Abstract
The emerging resistance to quinine jeopardizes the efficacy of a drug that has been used in the treatment of malaria for several centuries. To identify factors contributing to differential quinine responses in the human malaria parasite Plasmodium falciparum, we have conducted comparative quantitative trait locus analyses on the susceptibility to quinine and also its stereoisomer quinidine, and on the initial and steady-state intracellular drug accumulation levels in the F1 progeny of a genetic cross. These data, together with genetic screens of field isolates and laboratory strains associated differential quinine and quinidine responses with mutated pfcrt, a segment on chromosome 13, and a novel candidate gene, termed MAL7P1.19 (encoding a HECT ubiquitin ligase). Despite a strong likelihood of association, episomal transfections demonstrated a role for the HECT ubiquitin-protein ligase in quinine and quinidine sensitivity in only a subset of genetic backgrounds, and here the changes in IC50 values were moderate (approximately 2-fold). These data show that quinine responsiveness is a complex genetic trait with multiple alleles playing a role and that more experiments are needed to unravel the role of the contributing factors. Quinine, a natural product from cinchona bark, has been used in the treatment of malaria for centuries. Unfortunately, a progressive loss in responsiveness of the human malaria parasite Plasmodium falciparum to quinine has been observed, particularly in Southeast Asia, where cases of quinine treatment failure regularly occur. To better understand how P. falciparum defends itself against the cytotoxic activity of quinine, we have conducted comparative linkage analyses in the F1 progeny of a genetic cross where we assessed the susceptibility and the amount of intracellular accumulation of quinine and of its stereoisomer quinidine. These data identified a novel candidate gene encoding a HECT ubiquitin-protein ligase that might contribute to altered quinine responsiveness. The identification of this novel gene might improve the surveillance of quinine-resistant malaria parasites in the field and aid the preservation of this valuable antimalarial drug.
Collapse
Affiliation(s)
- Cecilia P. Sanchez
- Department of Infectious Diseases, Parasitology, Universitätsklinikum Heidelberg, Heidelberg, Germany
| | - Chia-Hao Liu
- Department of Infectious Diseases, Parasitology, Universitätsklinikum Heidelberg, Heidelberg, Germany
| | - Sybille Mayer
- Department of Infectious Diseases, Parasitology, Universitätsklinikum Heidelberg, Heidelberg, Germany
| | - Astutiati Nurhasanah
- Department of Infectious Diseases, Parasitology, Universitätsklinikum Heidelberg, Heidelberg, Germany
- Laboratory for the Development of Agroindustrial and Biomedical Technology (LAPTIAB), Tangerang Selatan, Indonesia
| | - Marek Cyrklaff
- Department of Infectious Diseases, Parasitology, Universitätsklinikum Heidelberg, Heidelberg, Germany
| | - Jianbing Mu
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Michael T. Ferdig
- The Eck Institute for Global Health, Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Wilfred D. Stein
- Biological Chemistry, Silberman Institute of Life Sciences, Hebrew University of Jerusalem, Givat Ram, Jerusalem, Israel
| | - Michael Lanzer
- Department of Infectious Diseases, Parasitology, Universitätsklinikum Heidelberg, Heidelberg, Germany
- * E-mail:
| |
Collapse
|
25
|
Ariey F, Witkowski B, Amaratunga C, Beghain J, Langlois AC, Khim N, Kim S, Duru V, Bouchier C, Ma L, Lim P, Leang R, Duong S, Sreng S, Suon S, Chuor CM, Bout DM, Ménard S, Rogers WO, Genton B, Fandeur T, Miotto O, Ringwald P, Le Bras J, Berry A, Barale JC, Fairhurst RM, Benoit-Vical F, Mercereau-Puijalon O, Ménard D. A molecular marker of artemisinin-resistant Plasmodium falciparum malaria. Nature 2014; 505:50-5. [PMID: 24352242 PMCID: PMC5007947 DOI: 10.1038/nature12876] [Citation(s) in RCA: 1490] [Impact Index Per Article: 135.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 11/12/2013] [Indexed: 12/24/2022]
Abstract
Plasmodium falciparum resistance to artemisinin derivatives in southeast Asia threatens malaria control and elimination activities worldwide. To monitor the spread of artemisinin resistance, a molecular marker is urgently needed. Here, using whole-genome sequencing of an artemisinin-resistant parasite line from Africa and clinical parasite isolates from Cambodia, we associate mutations in the PF3D7_1343700 kelch propeller domain ('K13-propeller') with artemisinin resistance in vitro and in vivo. Mutant K13-propeller alleles cluster in Cambodian provinces where resistance is prevalent, and the increasing frequency of a dominant mutant K13-propeller allele correlates with the recent spread of resistance in western Cambodia. Strong correlations between the presence of a mutant allele, in vitro parasite survival rates and in vivo parasite clearance rates indicate that K13-propeller mutations are important determinants of artemisinin resistance. K13-propeller polymorphism constitutes a useful molecular marker for large-scale surveillance efforts to contain artemisinin resistance in the Greater Mekong Subregion and prevent its global spread.
Collapse
Affiliation(s)
- Frédéric Ariey
- 1] Institut Pasteur, Parasite Molecular Immunology Unit, 75724 Paris Cedex 15, France [2] Centre National de la Recherche Scientifique, Unité de Recherche Associée 2581, 75724 Paris Cedex 15, France [3] Institut Pasteur, Genetics and Genomics of Insect Vectors Unit, 75724 Paris Cedex 15, France (F.A.); Institut Pasteur, Functional Genetics of Infectious Diseases Unit, 75724 Paris Cedex 15, France (J.B.); Centre de Physiopathologie de Toulouse-Purpan, Institut National de la Santé et de la Recherche Médicale UMR1043, Centre National de la Recherche Scientifique UMR5282, Université Toulouse III, 31024 Toulouse Cedex 3, France Institut Pasteur, Unité de Biologie et Génétique du Paludisme, Team Malaria Targets and Drug Development, 75724 Paris Cedex 15, France (J.-C.B.)
| | - Benoit Witkowski
- Institut Pasteur du Cambodge, Malaria Molecular Epidemiology Unit, Phnom Penh, Cambodia
| | - Chanaki Amaratunga
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Johann Beghain
- 1] Institut Pasteur, Parasite Molecular Immunology Unit, 75724 Paris Cedex 15, France [2] Centre National de la Recherche Scientifique, Unité de Recherche Associée 2581, 75724 Paris Cedex 15, France [3] Institut Pasteur, Genetics and Genomics of Insect Vectors Unit, 75724 Paris Cedex 15, France (F.A.); Institut Pasteur, Functional Genetics of Infectious Diseases Unit, 75724 Paris Cedex 15, France (J.B.); Centre de Physiopathologie de Toulouse-Purpan, Institut National de la Santé et de la Recherche Médicale UMR1043, Centre National de la Recherche Scientifique UMR5282, Université Toulouse III, 31024 Toulouse Cedex 3, France Institut Pasteur, Unité de Biologie et Génétique du Paludisme, Team Malaria Targets and Drug Development, 75724 Paris Cedex 15, France (J.-C.B.)
| | - Anne-Claire Langlois
- 1] Institut Pasteur, Parasite Molecular Immunology Unit, 75724 Paris Cedex 15, France [2] Centre National de la Recherche Scientifique, Unité de Recherche Associée 2581, 75724 Paris Cedex 15, France
| | - Nimol Khim
- Institut Pasteur du Cambodge, Malaria Molecular Epidemiology Unit, Phnom Penh, Cambodia
| | - Saorin Kim
- Institut Pasteur du Cambodge, Malaria Molecular Epidemiology Unit, Phnom Penh, Cambodia
| | - Valentine Duru
- Institut Pasteur du Cambodge, Malaria Molecular Epidemiology Unit, Phnom Penh, Cambodia
| | - Christiane Bouchier
- Institut Pasteur, Plate-forme Génomique, Département Génomes et Génétique, 75724 Paris Cedex 15, France
| | - Laurence Ma
- Institut Pasteur, Plate-forme Génomique, Département Génomes et Génétique, 75724 Paris Cedex 15, France
| | - Pharath Lim
- 1] Institut Pasteur du Cambodge, Malaria Molecular Epidemiology Unit, Phnom Penh, Cambodia [2] Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA [3] National Center for Parasitology, Entomology and Malaria Control, Phnom Penh, Cambodia
| | - Rithea Leang
- National Center for Parasitology, Entomology and Malaria Control, Phnom Penh, Cambodia
| | - Socheat Duong
- National Center for Parasitology, Entomology and Malaria Control, Phnom Penh, Cambodia
| | - Sokunthea Sreng
- National Center for Parasitology, Entomology and Malaria Control, Phnom Penh, Cambodia
| | - Seila Suon
- National Center for Parasitology, Entomology and Malaria Control, Phnom Penh, Cambodia
| | - Char Meng Chuor
- National Center for Parasitology, Entomology and Malaria Control, Phnom Penh, Cambodia
| | - Denis Mey Bout
- SSA WHO, Drug Monitoring in Cambodia, National Center for Parasitology, Entomology and Malaria Control, Phnom Penh, Cambodia
| | - Sandie Ménard
- 1] Service de Parasitologie et Mycologie, Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse Cedex 9, France [2] Institut Pasteur, Genetics and Genomics of Insect Vectors Unit, 75724 Paris Cedex 15, France (F.A.); Institut Pasteur, Functional Genetics of Infectious Diseases Unit, 75724 Paris Cedex 15, France (J.B.); Centre de Physiopathologie de Toulouse-Purpan, Institut National de la Santé et de la Recherche Médicale UMR1043, Centre National de la Recherche Scientifique UMR5282, Université Toulouse III, 31024 Toulouse Cedex 3, France Institut Pasteur, Unité de Biologie et Génétique du Paludisme, Team Malaria Targets and Drug Development, 75724 Paris Cedex 15, France (J.-C.B.)
| | | | - Blaise Genton
- Swiss Tropical and Public Health Institute, 4051 Basel, Switzerland
| | - Thierry Fandeur
- 1] Institut Pasteur, Parasite Molecular Immunology Unit, 75724 Paris Cedex 15, France [2] Institut Pasteur du Cambodge, Malaria Molecular Epidemiology Unit, Phnom Penh, Cambodia
| | - Olivo Miotto
- 1] MRC Centre for Genomics and Global Health, University of Oxford, Oxford OX3 7BN, UK [2] Mahidol-Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok 10400, Thailand [3] Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1SA, UK
| | - Pascal Ringwald
- Global Malaria Program, World Health Organization, 1211 Geneva, Switzerland
| | - Jacques Le Bras
- Centre National de Référence du Paludisme, CHU Bichat-Claude Bernard, APHP, PRES Sorbonne Paris Cité, 75018 Paris, France
| | - Antoine Berry
- 1] Service de Parasitologie et Mycologie, Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse Cedex 9, France [2] Institut Pasteur, Genetics and Genomics of Insect Vectors Unit, 75724 Paris Cedex 15, France (F.A.); Institut Pasteur, Functional Genetics of Infectious Diseases Unit, 75724 Paris Cedex 15, France (J.B.); Centre de Physiopathologie de Toulouse-Purpan, Institut National de la Santé et de la Recherche Médicale UMR1043, Centre National de la Recherche Scientifique UMR5282, Université Toulouse III, 31024 Toulouse Cedex 3, France Institut Pasteur, Unité de Biologie et Génétique du Paludisme, Team Malaria Targets and Drug Development, 75724 Paris Cedex 15, France (J.-C.B.)
| | - Jean-Christophe Barale
- 1] Institut Pasteur, Parasite Molecular Immunology Unit, 75724 Paris Cedex 15, France [2] Centre National de la Recherche Scientifique, Unité de Recherche Associée 2581, 75724 Paris Cedex 15, France [3] Institut Pasteur, Genetics and Genomics of Insect Vectors Unit, 75724 Paris Cedex 15, France (F.A.); Institut Pasteur, Functional Genetics of Infectious Diseases Unit, 75724 Paris Cedex 15, France (J.B.); Centre de Physiopathologie de Toulouse-Purpan, Institut National de la Santé et de la Recherche Médicale UMR1043, Centre National de la Recherche Scientifique UMR5282, Université Toulouse III, 31024 Toulouse Cedex 3, France Institut Pasteur, Unité de Biologie et Génétique du Paludisme, Team Malaria Targets and Drug Development, 75724 Paris Cedex 15, France (J.-C.B.)
| | - Rick M Fairhurst
- 1] Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA [2]
| | - Françoise Benoit-Vical
- 1] Centre National de la Recherche Scientifique, Laboratoire de Chimie de Coordination UPR8241, 31077 Toulouse Cedex 4, France [2] Université de Toulouse, UPS, Institut National Polytechnique de Toulouse, 31077 Toulouse Cedex 4, France [3]
| | - Odile Mercereau-Puijalon
- 1] Institut Pasteur, Parasite Molecular Immunology Unit, 75724 Paris Cedex 15, France [2] Centre National de la Recherche Scientifique, Unité de Recherche Associée 2581, 75724 Paris Cedex 15, France [3]
| | - Didier Ménard
- 1] Institut Pasteur du Cambodge, Malaria Molecular Epidemiology Unit, Phnom Penh, Cambodia [2]
| |
Collapse
|
26
|
Elbadry MA, Existe A, Victor YS, Memnon G, Fukuda M, Dame JB, Yowell CA, Okech BA. Survey of Plasmodium falciparum multidrug resistance-1 and chloroquine resistance transporter alleles in Haiti. Malar J 2013; 12:426. [PMID: 24252305 PMCID: PMC3879102 DOI: 10.1186/1475-2875-12-426] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2013] [Accepted: 10/25/2013] [Indexed: 12/04/2022] Open
Abstract
Background In Haiti where chloroquine (CQ) is widely used for malaria treatment, reports of resistance are scarce. However, recent identification of CQ resistance genotypes in one site is suggestive of an emerging problem. Additional studies are needed to evaluate genetic mutations associated with CQ resistance, especially in the Plasmodium falciparum multi-drug resistance-1 gene (pfmdr1) while expanding the already available information on P. falciparum CQ transporter gene (pfcrt) in Haiti. Methods Blood samples were collected on Whatman filter cards (FTA) from eight clinics spread across Haiti. Following the confirmation of P. falciparum in the samples, PCR protocols were used to amplify regions of pfmdr1and pfcrt codons of interest, (86, 184, 1034, 1042, and 1246) and (72-76), respectively. Sequencing and site-specific restriction enzyme digestions were used to analyse these DNA fragments for the presence of single nucleotide polymorphisms (SNPs) known to confer resistance to anti-malarial drugs. Results P. falciparum infection was confirmed in160 samples by amplifying a segment of the P. falciparum 18S small subunit ribosomal RNA gene (pfssurrna). The sequence of pfmdr1 in 54 of these samples was determined between codons 86,184 codons 1034, 1042 and 1246. No sequence differences from that of the NF54 clone 3D7 were found among the 54 samples except at codon 184, where a non-silent mutation was found in all samples predicted to alter the amino acid sequence replacing tyrosine with phenylalanine (Y184F). This altered sequence was also confirmed by restriction enzyme digestion. The sequence of pfmdr1 at codons 86, 184, 1034 and 1042 encoded the NFSN haplotype. The sequence of pfcrt codons 72-76 from 79 samples was determined and found to encode CVMNK, consistent with a CQ sensitive genotype. Conclusion The presence of the Y184F mutation in pfmdr1 of P. falciparum parasites in Haiti may have implications for resistance to antimalarial drugs. The absence of mutation in pfcrt at codon 76 among 79 isolates tested suggests that sensitivity to CQ in Haiti remains common. Wide-spread screening of the pfmdr1 and pfcrt especially among patients experiencing treatment failure may be a useful tool in early detection of the emergence of antimalarial drug resistance in Haiti.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Bernard A Okech
- Department of Environmental and Global Health, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
27
|
Egan TJ, Kuter D. Dual-functioning antimalarials that inhibit the chloroquine-resistance transporter. Future Microbiol 2013; 8:475-89. [PMID: 23534360 PMCID: PMC7099626 DOI: 10.2217/fmb.13.18] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Malaria remains a major international health challenge. Resistance to a number of existing drugs and evidence of the emergence of artemisinin resistance has emphasized the need for new antimalarials. A new approach has been the preparation of dual-function compounds that include a chloroquine-like antimalarial group and a group that resembles a chloroquine chemosensitizer. This article reviews the recent discovery of such dual-function antimalarials that are proposed to target both hemozoin formation and the chloroquine resistance transporter, PfCRT. These are discussed in relation to the mechanism of action of 4-aminoquinolines, chloroquine resistance and resistance reversal.
Collapse
Affiliation(s)
- Timothy J Egan
- Department of Chemistry, University of Cape Town, Private Bag, Rondebosch 7701, South Africa.
| | | |
Collapse
|
28
|
Patel SK, George LB, Prasanth Kumar S, Highland HN, Jasrai YT, Pandya HA, Desai KR. A Computational Approach towards the Understanding of Plasmodium falciparum Multidrug Resistance Protein 1. ISRN BIOINFORMATICS 2013; 2013:437168. [PMID: 25937947 PMCID: PMC4393060 DOI: 10.1155/2013/437168] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Accepted: 07/02/2013] [Indexed: 11/17/2022]
Abstract
The emergence of drug resistance in Plasmodium falciparum tremendously affected the chemotherapy worldwide while the intense distribution of chloroquine-resistant strains in most of the endemic areas added more complications in the treatment of malaria. The situation has even worsened by the lack of molecular mechanism to understand the resistance conferred by Plasmodia species. Recent studies have suggested the association of antimalarial resistance with P. falciparum multidrug resistance protein 1 (PfMDR1), an ATP-binding cassette (ABC) transporter and a homologue of human P-glycoprotein 1 (P-gp1). The present study deals about the development of PfMDR1 computational model and the model of substrate transport across PfMDR1 with insights derived from conformations relative to inward- and outward-facing topologies that switch on/off the transportation system. Comparison of ATP docked positions and its structural motif binding properties were found to be similar among other ATPases, and thereby contributes to NBD domains dimerization, a unique structural agreement noticed in Mus musculus Pgp and Escherichia coli MDR transporter homolog (MsbA). The interaction of leading antimalarials and phytochemicals within the active pocket of both wild-type and mutant-type PfMDR1 demonstrated the mode of binding and provided insights of less binding affinity thereby contributing to parasite's resistance mechanism.
Collapse
Affiliation(s)
- Saumya K. Patel
- Department of Bioinformatics, Applied Botany Centre (ABC), University School of Sciences, Gujarat University, Ahmedabad 380009, India
- Department of Zoology, Biomedical Technology and Human Genetics, University School of Sciences, Gujarat University, Ahmedabad 380009, India
| | - Linz-Buoy George
- Department of Zoology, Biomedical Technology and Human Genetics, University School of Sciences, Gujarat University, Ahmedabad 380009, India
| | - Sivakumar Prasanth Kumar
- Department of Bioinformatics, Applied Botany Centre (ABC), University School of Sciences, Gujarat University, Ahmedabad 380009, India
| | - Hyacinth N. Highland
- Department of Zoology, Biomedical Technology and Human Genetics, University School of Sciences, Gujarat University, Ahmedabad 380009, India
| | - Yogesh T. Jasrai
- Department of Bioinformatics, Applied Botany Centre (ABC), University School of Sciences, Gujarat University, Ahmedabad 380009, India
| | - Himanshu A. Pandya
- Department of Bioinformatics, Applied Botany Centre (ABC), University School of Sciences, Gujarat University, Ahmedabad 380009, India
| | - Ketaki R. Desai
- Department of Zoology, Biomedical Technology and Human Genetics, University School of Sciences, Gujarat University, Ahmedabad 380009, India
| |
Collapse
|
29
|
Brunner R, Ng CL, Aissaoui H, Akabas MH, Boss C, Brun R, Callaghan PS, Corminboeuf O, Fidock DA, Frame IJ, Heidmann B, Le Bihan A, Jenö P, Mattheis C, Moes S, Müller IB, Paguio M, Roepe PD, Siegrist R, Voss T, Welford RWD, Wittlin S, Binkert C. UV-triggered affinity capture identifies interactions between the Plasmodium falciparum multidrug resistance protein 1 (PfMDR1) and antimalarial agents in live parasitized cells. J Biol Chem 2013; 288:22576-83. [PMID: 23754276 DOI: 10.1074/jbc.m113.453159] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
A representative of a new class of potent antimalarials with an unknown mode of action was recently described. To identify the molecular target of this class of antimalarials, we employed a photo-reactive affinity capture method to find parasite proteins specifically interacting with the capture compound in living parasitized cells. The capture reagent retained the antimalarial properties of the parent molecule (ACT-213615) and accumulated within parasites. We identified several proteins interacting with the capture compound and established a functional interaction between ACT-213615 and PfMDR1. We surmise that PfMDR1 may play a role in the antimalarial activity of the piperazine-containing compound ACT-213615.
Collapse
Affiliation(s)
- Ralf Brunner
- Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, CH-4002 Basel, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Antao T, Hastings I. Policy options for deploying anti-malarial drugs in endemic countries: a population genetics approach. Malar J 2012; 11:422. [PMID: 23244624 PMCID: PMC3546853 DOI: 10.1186/1475-2875-11-422] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Accepted: 12/08/2012] [Indexed: 11/30/2022] Open
Abstract
Background Anti-malarial drugs are constantly exposed to the threat of evolving drug resistance so good stewardship of existing therapy is an essential component of public health policy. However, the widespread availability of numerous different drugs through informal providers could undermine official drug deployment policies. A policy of multiple first-line therapy (MFT) is compared with the conventional policy of sequential drug deployment, i.e., where one drug is used until resistance evolves and then replaced by the next drug in the sequence. Methods Population genetic models of drug resistance are used to make the comparison; this methodology explicitly tracks the genetics of drug resistance (including, importantly, recombination in the sexual stage, intrahost dynamics, and direction of linkage disequilibrium). Results A policy of MFT outlasts sequential application providing drug usages are low to moderate, and appears not to drive widespread multi-drug resistance. Inadequate dosing is an even more potent driver of drug resistance than the MFT/sequential policy decision. Conclusions The provision of MFT as a deliberate policy can be encouraged provided overall treatment rates are low or moderate (less than around half of malaria infections are treated) and the ad hoc provision of MFT through the private sector may be tolerated. This must be fully supported by education to ensure people take adequate doses of each of the drugs.
Collapse
Affiliation(s)
- Tiago Antao
- Liverpool School of Tropical Medicine, Pembroke Place, Liverpool L3 5QA, UK.
| | | |
Collapse
|
31
|
PfCRT and its role in antimalarial drug resistance. Trends Parasitol 2012; 28:504-14. [PMID: 23020971 DOI: 10.1016/j.pt.2012.08.002] [Citation(s) in RCA: 196] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Revised: 08/09/2012] [Accepted: 08/13/2012] [Indexed: 12/15/2022]
Abstract
Plasmodium falciparum resistance to chloroquine, the former gold standard antimalarial drug, is mediated primarily by mutant forms of the chloroquine resistance transporter (PfCRT). These mutations impart upon PfCRT the ability to efflux chloroquine from the intracellular digestive vacuole, the site of drug action. Recent studies reveal that PfCRT variants can also affect parasite fitness, protect immature gametocytes against chloroquine action, and alter P. falciparum susceptibility to current first-line therapies. These results highlight the need to be vigilant in screening for the appearance of novel pfcrt alleles that could contribute to new multi-drug resistance phenotypes.
Collapse
|
32
|
Cui L, Wang Z, Miao J, Miao M, Chandra R, Jiang H, Su XZ, Cui L. Mechanisms of in vitro resistance to dihydroartemisinin in Plasmodium falciparum. Mol Microbiol 2012; 86:111-28. [PMID: 22812578 DOI: 10.1111/j.1365-2958.2012.08180.x] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The recent reports of artemisinin (ART) resistance in the Thai-Cambodian border area raise a serious concern on the long-term efficacy of ARTs. To elucidate the resistance mechanisms, we performed in vitro selection with dihydroartemisinin (DHA) and obtained two parasite clones from Dd2 with more than 25-fold decrease in susceptibility to DHA. The DHA-resistant clones were more tolerant of stressful growth conditions and more resistant to several commonly used antimalarial drugs than Dd2. The result is worrisome as many of the drugs are currently used as ART partners in malaria control. This study showed that the DHA resistance is not limited to ring stage, but also occurred in trophozoites and schizonts. Microarray and biochemical analyses revealed pfmdr1 amplification, elevation of the antioxidant defence network, and increased expression of many chaperones in the DHA-resistant parasites. Without drug pressure, the DHA-resistant parasites reverted to sensitivity in approximately 8 weeks, accompanied by de-amplification of pfmdr1 and reduced antioxidant activities. The parallel decrease and increase in pfmdr1 copy number and antioxidant activity and the up and down of DHA sensitivity strongly suggest that pfmdr1 and antioxidant defence play a role in in vitro resistance to DHA, providing potential molecular markers for ART resistance.
Collapse
Affiliation(s)
- Long Cui
- Department of Entomology, The Pennsylvania State University, University Park, PA 16802, USA
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Pillai DR, Lau R, Khairnar K, Lepore R, Via A, Staines HM, Krishna S. Artemether resistance in vitro is linked to mutations in PfATP6 that also interact with mutations in PfMDR1 in travellers returning with Plasmodium falciparum infections. Malar J 2012; 11:131. [PMID: 22540925 PMCID: PMC3422158 DOI: 10.1186/1475-2875-11-131] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Accepted: 04/27/2012] [Indexed: 11/28/2022] Open
Abstract
Background Monitoring resistance phenotypes for Plasmodium falciparum, using in vitro growth assays, and relating findings to parasite genotype has proved particularly challenging for the study of resistance to artemisinins. Methods Plasmodium falciparum isolates cultured from 28 returning travellers diagnosed with malaria were assessed for sensitivity to artemisinin, artemether, dihydroartemisinin and artesunate and findings related to mutations in pfatp6 and pfmdr1. Results Resistance to artemether in vitro was significantly associated with a pfatp6 haplotype encoding two amino acid substitutions (pfatp6 A623E and S769N; (mean IC50 (95% CI) values of 8.2 (5.7 – 10.7) for A623/S769 versus 623E/769 N 13.5 (9.8 – 17.3) nM with a mean increase of 65%; p = 0.012). Increased copy number of pfmdr1 was not itself associated with increased IC50 values for artemether, but when interactions between the pfatp6 haplotype and increased copy number of pfmdr1 were examined together, a highly significant association was noted with IC50 values for artemether (mean IC50 (95% CI) values of 8.7 (5.9 – 11.6) versus 16.3 (10.7 – 21.8) nM with a mean increase of 87%; p = 0.0068). Previously described SNPs in pfmdr1 are also associated with differences in sensitivity to some artemisinins. Conclusions These findings were further explored in molecular modelling experiments that suggest mutations in pfatp6 are unlikely to affect differential binding of artemisinins at their proposed site, whereas there may be differences in such binding associated with mutations in pfmdr1. Implications for a hypothesis that artemisinin resistance may be exacerbated by interactions between PfATP6 and PfMDR1 and for epidemiological studies to monitor emerging resistance are discussed.
Collapse
Affiliation(s)
- Dylan R Pillai
- Centre for Infection and Immunity, Division of Clinical Sciences, St, George's, University of London, London SW17 0RE, UK
| | | | | | | | | | | | | |
Collapse
|
34
|
Mullié C, Jonet A, Desgrouas C, Taudon N, Sonnet P. Differences in anti-malarial activity of 4-aminoalcohol quinoline enantiomers and investigation of the presumed underlying mechanism of action. Malar J 2012; 11:65. [PMID: 22401346 PMCID: PMC3314553 DOI: 10.1186/1475-2875-11-65] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Accepted: 03/08/2012] [Indexed: 11/10/2022] Open
Abstract
Background A better anti-malarial efficiency and lower neurotoxicity have been reported for mefloquine (MQ) (+)- enantiomer. However, the importance of stereoselectivity remains poorly understood as the anti-malarial activity of pure enantiomer MQ analogues has never been described. Building on these observations, a series of enantiopure 4-aminoalcohol quinoline derivatives has previously been synthesized to optimize the efficiency and reduce possible adverse effects. Their in vitro activity on Plasmodium falciparum W2 and 3D7 strains is reported here along with their inhibition of β-haematin formation and peroxidative degradation of haemin, two possible mechanisms of action of anti-malarial drugs. Results The (S)-enantiomers of this series of 4-aminoalcohol quinoline derivatives were found to be at least as effective as both chloroquine (CQ) and MQ. The derivative with a 5-carbon side-chain length was the more efficient on both P. falciparum strains. (R )-enantiomers displayed an activity decreased by 2 to 15-fold as compared to their (S) counterparts. The inhibition of β-haematin formation was significantly stronger with all tested compounds than with MQ, irrespective of the stereochemistry. Similarly, the inhibition of haemin peroxidation was significantly higher for both (S) and (R)-enantiomers of derivatives with a side-chain length of five or six carbons than for MQ and CQ. Conclusions The prominence of stereochemistry in the anti-malarial activity of 4-aminoalcohol quinoline derivatives is confirmed. The inhibition of β-haematin formation and haemin peroxidation can be put forward as presumed mechanisms of action but do not account for the stereoselectivity of action witnessed in vitro.
Collapse
Affiliation(s)
- Catherine Mullié
- Laboratoire des Glucides, UMR-CNRS 6219, UFR de Pharmacie, 1 rue des Louvels, 80037 Amiens Cedex 1, France.
| | | | | | | | | |
Collapse
|
35
|
Polymorphisms of the pfmdr1 but not the pfnhe-1 gene is associated with in vitro quinine sensitivity in Thai isolates of Plasmodium falciparum. Malar J 2012; 11:7. [PMID: 22221394 PMCID: PMC3287963 DOI: 10.1186/1475-2875-11-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Revised: 07/12/2011] [Accepted: 01/05/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The emergence of Plasmodium falciparum resistance to most currently used anti-malarial drugs is a major problem in malaria control along the Thai-Myanmar and Thai-Cambodia borders. Quinine (QN) with tetracycline/doxycycline has been used as the second-line treatment for uncomplicated falciparum malaria. In addition, QN monotherapy has been the first-line treatment for falciparum malaria in pregnant women. However, reduced in vitro and in vivo responses to QN have been reported. To date, a few genetic markers for QN resistance have been proposed including Plasmodium falciparum chloroquine resistance transporter (pfcrt), P. falciparum multidrug resistance 1 (pfmdr1), and P. falciparum Na+/H+ exchanger (pfnhe-1). This study was to investigate the role of the pfmdr1 and pfnhe-1 gene on in vitro QN sensitivity in Thai isolates of P. falciparum. METHODS Eighty-five Thai isolates of P. falciparum from the Thai-Myanmar and Thai-Cambodia borders from 2003-2008 were determined for in vitro QN sensitivity using radioisotopic assay. Polymorphisms of the pfmdr1 and pfnhe-1 gene were determined by PCR-RFLP and sequence analysis. Associations between the in vitro QN sensitivity and the polymorphisms of the pfmdr1 and pfnhe-1 gene were evaluated. RESULTS The mean QN IC50 was 202.8 nM (range 25.7-654.4 nM). Only four isolates were QN resistant when the IC50 of >500 nM was used as the cut-off point. Significant associations were found between the pfmdr1 mutations at codons N86Y and N1042D and in vitro QN sensitivity. However, no associations with the number of DNNND, DDNNNDNHNDD, and NHNDNHNNDDD repeats in the microsatellite ms4760 of the pfnhe-1 gene were identified. CONCLUSION Data from the present study put doubt regarding the pfnhe-1 gene as to whether it could be used as the suitable marker for QN resistance in Thailand. In contrast, it confirms the influence of the pfmdr1 gene on in vitro QN sensitivity.
Collapse
|
36
|
Ferreira PE, Holmgren G, Veiga MI, Uhlén P, Kaneko A, Gil JP. PfMDR1: mechanisms of transport modulation by functional polymorphisms. PLoS One 2011; 6:e23875. [PMID: 21912647 PMCID: PMC3164660 DOI: 10.1371/journal.pone.0023875] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2011] [Accepted: 07/27/2011] [Indexed: 01/22/2023] Open
Abstract
ATP-Binding Cassette (ABC) transporters are efflux pumps frequently associated with multidrug resistance in many biological systems, including malaria. Antimalarial drug-resistance involves an ABC transporter, PfMDR1, a homologue of P-glycoprotein in humans. Twenty years of research have shown that several single nucleotide polymorphisms in pfmdr1 modulate in vivo and/or in vitro drug susceptibility. The underlying physiological mechanism of the effect of these mutations remains unclear. Here we develop structural models for PfMDR1 in different predicted conformations, enabling the study of transporter motion. Such analysis of functional polymorphisms allows determination of their potential role in transport and resistance. The bacterial MsbA ABC pump is a PfMDR1 homologue. MsbA crystals in different conformations were used to create PfMDR1 models with Modeller software. Sequences were aligned with ClustalW and analysed by Ali2D revealing a high level of secondary structure conservation. To validate a potential drug binding pocket we performed antimalarial docking simulations. Using aminoquinoline as probe drugs in PfMDR1 mutated parasites we evaluated the physiology underlying the mechanisms of resistance mediated by PfMDR1 polymorphisms. We focused on the analysis of well known functional polymorphisms in PfMDR1 amino acid residues 86, 184, 1034, 1042 and 1246. Our structural analysis suggested the existence of two different biophysical mechanisms of PfMDR1 drug resistance modulation. Polymorphisms in residues 86/184/1246 act by internal allosteric modulation and residues 1034 and 1042 interact directly in a drug pocket. Parasites containing mutated PfMDR1 variants had a significant altered aminoquinoline susceptibility that appears to be dependent on the aminoquinoline lipophobicity characteristics as well as vacuolar efflux by PfCRT. We previously described the in vivo selection of PfMDR1 polymorphisms under antimalarial drug pressure. Now, together with recent PfMDR1 functional reports, we contribute to the understanding of the specific structural role of these polymorphisms in parasite antimalarial drug response.
Collapse
|
37
|
Differential drug efflux or accumulation does not explain variation in the chloroquine response of Plasmodium falciparum strains expressing the same isoform of mutant PfCRT. Antimicrob Agents Chemother 2011; 55:2310-8. [PMID: 21343459 DOI: 10.1128/aac.01167-10] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Mutant forms of the Plasmodium falciparum chloroquine resistance transporter (PfCRT) mediate chloroquine resistance by effluxing the drug from the parasite's digestive vacuole, the acidic organelle in which chloroquine exerts its parasiticidal effect. However, different parasites bearing the same mutant form of PfCRT can vary substantially in their chloroquine susceptibility. Here, we have investigated the biochemical basis for the difference in chloroquine response among transfectant parasite lines having different genetic backgrounds but bearing the same mutant form of PfCRT. Despite showing significant differences in their chloroquine susceptibility, all lines with the mutant PfCRT showed a similar chloroquine-induced H+ leak from the digestive vacuole, indicative of similar rates of PfCRT-mediated chloroquine efflux. Furthermore, all lines showed similarly reduced levels of drug accumulation. Factors other than chloroquine efflux and accumulation therefore influence the susceptibility to this drug in parasites expressing mutant PfCRT. Furthermore, in some but not all strains bearing mutant PfCRT, the 50% inhibitory concentration (IC50) for chloroquine and the degree of resistance compared to that of recombinant control parasites varied with the length of the parasite growth assays. In these parasites, the 50% inhibitory concentration for chloroquine measured in 72- or 96-h assays was significantly lower than that measured in 48-h assays. This highlights the importance of considering the first- and second-cycle activities of chloroquine in future studies of parasite susceptibility to this drug.
Collapse
|
38
|
Genetic predisposition favors the acquisition of stable artemisinin resistance in malaria parasites. Antimicrob Agents Chemother 2010; 55:50-5. [PMID: 21041511 DOI: 10.1128/aac.00916-10] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The emergence of artemisinin-resistant Plasmodium falciparum malaria jeopardizes efforts to control this infectious disease. To identify factors contributing to reduced artemisinin susceptibility, we have employed a classical genetic approach by analyzing artemisinin responses in the F1 progeny of a genetic cross. Our data show that reduced artemisinin susceptibility is a multifactorial trait, with pfmdr1 and two additional loci (on chromosomes 12 and 13) contributing to it. We further show that the different artemisinin susceptibilities of the progeny strains affect their responses to selection with increasing concentrations of artemisinin. Stable, high-level in vitro artemisinin resistance rapidly arose in those parasites that were the least artemisinin susceptible among the F1 progeny, whereas progeny that were highly artemisinin susceptible did not acquire stable artemisinin resistance. These data suggest that genetic predisposition favors the acquisition of high-level artemisinin resistance. In vitro-induced artemisinin resistance did not result in cross-resistance to artesunate or artemether, suggesting that resistance to one derivative does not necessarily render the entire drug class ineffective.
Collapse
|
39
|
Patel JJ, Thacker D, Tan JC, Pleeter P, Checkley L, Gonzales JM, Deng B, Roepe PD, Cooper RA, Ferdig MT. Chloroquine susceptibility and reversibility in a Plasmodium falciparum genetic cross. Mol Microbiol 2010; 78:770-87. [PMID: 20807203 DOI: 10.1111/j.1365-2958.2010.07366.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Mutations in the Plasmodium falciparum chloroquine (CQ) resistance transporter (PfCRT) are major determinants of verapamil (VP)-reversible CQ resistance (CQR). In the presence of mutant PfCRT, additional genes contribute to the wide range of CQ susceptibilities observed. It is not known if these genes influence mechanisms of chemosensitization by CQR reversal agents. Using quantitative trait locus (QTL) mapping of progeny clones from the HB3 × Dd2 cross, we show that the P. falciparum multidrug resistance gene 1 (pfmdr1) interacts with the South-East Asia-derived mutant pfcrt haplotype to modulate CQR levels. A novel chromosome 7 locus is predicted to contribute with the pfcrt and pfmdr1 loci to influence CQR levels. Chemoreversal via a wide range of chemical structures operates through a direct pfcrt-based mechanism. Direct inhibition of parasite growth by these reversal agents is influenced by pfcrt mutations and additional loci. Direct labelling of purified recombinant PfMDR1 protein with a highly specific photoaffinity CQ analogue, and lack of competition for photolabelling by VP, supports our QTL predictions. We find no evidence that pfmdr1 copy number affects CQ response in the progeny; however, inheritance patterns indicate that an allele-specific interaction between pfmdr1 and pfcrt is part of the complex genetic background of CQR.
Collapse
Affiliation(s)
- Jigar J Patel
- The Eck Institute for Global Health, Department of Biological Sciences, University of Notre Dame, 205 Galvin Life Sciences, Notre Dame, IN 46556, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Chavchich M, Gerena L, Peters J, Chen N, Cheng Q, Kyle DE. Role of pfmdr1 amplification and expression in induction of resistance to artemisinin derivatives in Plasmodium falciparum. Antimicrob Agents Chemother 2010; 54:2455-64. [PMID: 20350946 PMCID: PMC2876417 DOI: 10.1128/aac.00947-09] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2009] [Revised: 09/08/2009] [Accepted: 03/17/2010] [Indexed: 01/07/2023] Open
Abstract
Artemisinin and its derivatives are the most rapidly acting and efficacious antimalarial drugs currently available. Although resistance to these drugs has not been documented, there is growing concern about the potential for resistance to develop. In this paper we report the selection of parasite resistance to artelinic acid (AL) and artemisinin (QHS) in vitro and the molecular changes that occurred during the selection. Exposure of three Plasmodium falciparum lines (W2, D6, and TM91C235) to AL resulted in decreases in parasite susceptibilities to AL and QHS, as well as to mefloquine, quinine, halofantrine, and lumefantrine. The changes in parasite susceptibility were accompanied by increases in the copy number, mRNA expression, and protein expression of the pfmdr1 gene in the resistant progenies of W2 and TM91C235 parasites but not in those of D6 parasites. No changes were detected in the coding sequences of the pfmdr1, pfcrt, pfatp6, pftctp, and pfubcth genes or in the expression levels of pfatp6 and pftctp. Our data demonstrate that P. falciparum lines have the capacity to develop resistance to artemisinin derivatives in vitro and that this resistance is achieved by multiple mechanisms, to include amplification and increased expression of pfmdr1, a mechanism that also confers resistance to mefloquine. This observation is of practical importance, because artemisinin drugs are often used in combination with mefloquine for the treatment of malaria.
Collapse
Affiliation(s)
- Marina Chavchich
- Australian Army Malaria Institute, Weary Dunlop Drive, Gallipoli Barracks, Enoggera, Q4051, Queensland Institute of Medical Research, PO Royal Brisbane Hospital, Herston, Q4029, Australia, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, Department of Global Health, College of Public Health, University of South Florida, 3720 Spectrum Blvd., Tampa, Florida 33612
| | - Lucia Gerena
- Australian Army Malaria Institute, Weary Dunlop Drive, Gallipoli Barracks, Enoggera, Q4051, Queensland Institute of Medical Research, PO Royal Brisbane Hospital, Herston, Q4029, Australia, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, Department of Global Health, College of Public Health, University of South Florida, 3720 Spectrum Blvd., Tampa, Florida 33612
| | - Jennifer Peters
- Australian Army Malaria Institute, Weary Dunlop Drive, Gallipoli Barracks, Enoggera, Q4051, Queensland Institute of Medical Research, PO Royal Brisbane Hospital, Herston, Q4029, Australia, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, Department of Global Health, College of Public Health, University of South Florida, 3720 Spectrum Blvd., Tampa, Florida 33612
| | - Nanhua Chen
- Australian Army Malaria Institute, Weary Dunlop Drive, Gallipoli Barracks, Enoggera, Q4051, Queensland Institute of Medical Research, PO Royal Brisbane Hospital, Herston, Q4029, Australia, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, Department of Global Health, College of Public Health, University of South Florida, 3720 Spectrum Blvd., Tampa, Florida 33612
| | - Qin Cheng
- Australian Army Malaria Institute, Weary Dunlop Drive, Gallipoli Barracks, Enoggera, Q4051, Queensland Institute of Medical Research, PO Royal Brisbane Hospital, Herston, Q4029, Australia, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, Department of Global Health, College of Public Health, University of South Florida, 3720 Spectrum Blvd., Tampa, Florida 33612
| | - Dennis E. Kyle
- Australian Army Malaria Institute, Weary Dunlop Drive, Gallipoli Barracks, Enoggera, Q4051, Queensland Institute of Medical Research, PO Royal Brisbane Hospital, Herston, Q4029, Australia, Division of Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, Department of Global Health, College of Public Health, University of South Florida, 3720 Spectrum Blvd., Tampa, Florida 33612
| |
Collapse
|
41
|
Pillai AD, Pain M, Solomon T, Bokhari AAB, Desai SA. A cell-based high-throughput screen validates the plasmodial surface anion channel as an antimalarial target. Mol Pharmacol 2010; 77:724-33. [PMID: 20101003 PMCID: PMC2872968 DOI: 10.1124/mol.109.062711] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2009] [Accepted: 01/20/2010] [Indexed: 01/30/2023] Open
Abstract
The plasmodial surface anion channel (PSAC) is an unusual small-conductance ion channel induced on erythrocytes infected with plasmodia, including parasites responsible for human malaria. Although broadly available inhibitors produce microscopic clearance of parasite cultures at high concentrations and suggest that PSAC is an antimalarial target, they have low affinity for the channel and may interfere with other parasite activities. To address these concerns, we developed a miniaturized assay for PSAC activity and carried out a high-throughput inhibitor screen. Approximately 70,000 compounds from synthetic and natural product libraries were screened, revealing inhibitors from multiple structural classes including two novel and potent heterocyclic scaffolds. Single-channel patch-clamp studies indicated that these compounds act directly on PSAC, further implicating a proposed role in transport of diverse solutes. A statistically significant correlation between channel inhibition and in vitro parasite killing by a family of compounds provided chemical validation of PSAC as a drug target. These new inhibitors should be important research tools and may be starting points for much-needed antimalarial drugs.
Collapse
Affiliation(s)
- Ajay D Pillai
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852-8132, USA
| | | | | | | | | |
Collapse
|
42
|
dos Santos SC, Tenreiro S, Palma M, Becker J, Sá-Correia I. Transcriptomic profiling of the Saccharomyces cerevisiae response to quinine reveals a glucose limitation response attributable to drug-induced inhibition of glucose uptake. Antimicrob Agents Chemother 2009; 53:5213-23. [PMID: 19805573 PMCID: PMC2786357 DOI: 10.1128/aac.00794-09] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2009] [Revised: 09/02/2009] [Accepted: 09/23/2009] [Indexed: 11/20/2022] Open
Abstract
Quinine has been employed in the treatment of malaria for centuries and is still used against severe Plasmodium falciparum malaria. However, its interactions with the parasite remain poorly understood and subject to debate. In this study, we used the Saccharomyces cerevisiae eukaryotic model to better understand quinine's mode of action and the mechanisms underlying the cell response to the drug. We obtained a transcriptomic profile of the yeast's early response to quinine, evidencing a marked activation of genes involved in the low-glucose response (e.g., CAT8, ADR1, MAL33, MTH1, and SNF3). We used a low inhibitory quinine concentration with no detectable effect on plasma membrane function, consistent with the absence of a general nutrient starvation response and suggesting that quinine-induced glucose limitation is a specific response. We have further shown that transport of [(14)C]glucose is inhibited by quinine, with kinetic data indicating competitive inhibition. Also, tested mutant strains deleted for genes encoding high- and low-affinity hexose transporters (HXT1 to HXT5, HXT8, and HXT10) exhibit resistance phenotypes, correlating with reduced levels of quinine accumulation in the mutants examined. These results suggest that the hexose transporters are facilitators of quinine uptake in S. cerevisiae, possibly through a competitive inhibition mechanism. Interestingly, P. falciparum is highly dependent on glucose uptake, which is mediated by the single-copy transporter PfHT1, a protein with high homology to yeast's hexose transporters. We propose that PfHT1 is an interesting candidate quinine target possibly involved in quinine import in P. falciparum, an uptake mechanism postulated in recent studies to occur through a still-unidentified importer(s).
Collapse
Affiliation(s)
- Sandra C. dos Santos
- Institute for Biotechnology and Bioengineering (IBB), Centre for Biological and Chemical Engineering, Instituto Superior Técnico, 1049-001 Lisbon, Portugal, Affymetrix Core Facility, Instituto Gulbenkian Ciência, Oeiras, Portugal
| | - Sandra Tenreiro
- Institute for Biotechnology and Bioengineering (IBB), Centre for Biological and Chemical Engineering, Instituto Superior Técnico, 1049-001 Lisbon, Portugal, Affymetrix Core Facility, Instituto Gulbenkian Ciência, Oeiras, Portugal
| | - Margarida Palma
- Institute for Biotechnology and Bioengineering (IBB), Centre for Biological and Chemical Engineering, Instituto Superior Técnico, 1049-001 Lisbon, Portugal, Affymetrix Core Facility, Instituto Gulbenkian Ciência, Oeiras, Portugal
| | - Jorg Becker
- Institute for Biotechnology and Bioengineering (IBB), Centre for Biological and Chemical Engineering, Instituto Superior Técnico, 1049-001 Lisbon, Portugal, Affymetrix Core Facility, Instituto Gulbenkian Ciência, Oeiras, Portugal
| | - Isabel Sá-Correia
- Institute for Biotechnology and Bioengineering (IBB), Centre for Biological and Chemical Engineering, Instituto Superior Técnico, 1049-001 Lisbon, Portugal, Affymetrix Core Facility, Instituto Gulbenkian Ciência, Oeiras, Portugal
| |
Collapse
|
43
|
Paguio MF, Cabrera M, Roepe PD. Chloroquine transport in Plasmodium falciparum. 2. Analysis of PfCRT-mediated drug transport using proteoliposomes and a fluorescent chloroquine probe. Biochemistry 2009; 48:9482-91. [PMID: 19725576 PMCID: PMC2778255 DOI: 10.1021/bi901035j] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Mutations in the PfCRT protein cause chloroquine resistance (CQR), and earlier studies from our laboratory using plasma membrane inside-out vesicles (ISOV) prepared from yeast expressing recombinant PfCRT [Zhang, H., et al. (2004) Biochemistry 43, 8290-8296] suggested that the putative transporter mediates downhill facilitated diffusion of charged chloroquine (CQ). However, more recent experiments with a fluorescent CQ probe (NBD-CQ) presented in the accompanying paper (DOI 10.1021/bi901034r ) indicated that the CQR phenotype in live parasites is associated with a reduced rate of ATP-dependent CQ uptake into the digestive vacuole (DV). An altered rate constant for uptake has multiple interpretations. To further investigate this phenomenon, PfCRT proteins found in chloroquine-sensitive (CQS) and CQR strains of Plasmodium falciparum were purified from yeast engineered to express "yeast optimized" pfcrt genes, reconstituted into proteoliposomes (PL), and efflux of NBD-CQ was measured from these PL. A membrane-impermeant quencher was used to distinguish intra-PL NBD-CQ from extra-PL NBD-CQ vs time as well as resolve initial rates and rate constants for efflux. Efflux was investigated at a range of NBD-CQ concentrations, in the presence vs absence of pH gradients (DeltapH) and transmembrane potentials (DeltaPsi). Explicit turnover numbers for apparent PfCRT-mediated transport were then calculated under these conditions. Our data are consistent with a model wherein PfCRT catalyzes electrochemically downhill diffusion of NBD-CQ out of the DV, in response to DeltaPsi or DeltapH, at a rate that can partially compete with the ATP-dependent uptake of NBD-CQ by CQS parasites described in the previous paper. These data allow us to propose a refined model for altered CQ accumulation in CQR malarial parasites.
Collapse
Affiliation(s)
- Michelle F. Paguio
- Department of Chemistry, Georgetown University, 37th and O Streets, NW Washington, DC 20057
| | - Mynthia Cabrera
- Department of Chemistry, Georgetown University, 37th and O Streets, NW Washington, DC 20057
| | - Paul D. Roepe
- Department of Chemistry, Georgetown University, 37th and O Streets, NW Washington, DC 20057
- Department of Biochemistry and Cellular & Molecular Biology, Georgetown University, 37th and O Streets, NW Washington, DC 20057
- Center for Infectious Diseases, Georgetown University, 37th and O Streets, NW Washington, DC 20057
| |
Collapse
|
44
|
Kavishe RA, van den Heuvel JMW, van de Vegte-Bolmer M, Luty AJF, Russel FGM, Koenderink JB. Localization of the ATP-binding cassette (ABC) transport proteins PfMRP1, PfMRP2, and PfMDR5 at the Plasmodium falciparum plasma membrane. Malar J 2009; 8:205. [PMID: 19715599 PMCID: PMC2744701 DOI: 10.1186/1475-2875-8-205] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2009] [Accepted: 08/28/2009] [Indexed: 11/10/2022] Open
Abstract
Background The spread of drug resistance has been a major obstacle to the control of malaria. The mechanisms underlying drug resistance in malaria seem to be complex and multigenic. The current literature on multiple drug resistance against anti-malarials has documented PfMDR1, an ATP-binding cassette (ABC) protein, as an important determinant of resistance. In the Plasmodium falciparum genome, there are several ABC transporters some of which could be putative drug transporting proteins. In order to understand the molecular mechanisms underlying drug resistance, characterization of these transporters is essential. The aim of this study was to characterize and localize putative ABC transporters. Methods In the plasmoDB database, 16 members of the P. falciparum ABC family can be identified, 11 of which are putative transport proteins. A phylogenetic analysis of the aligned NBDs of the PfABC genes was performed. Antibodies against PfMRP1 (PfABCC1), PfMRP2 (PfABCC2), and PfMDR5 (PfABCB5) were generated, affinity purified and used in immunocytochemistry to localize the proteins in the asexual stages of the parasite. Results The ABC family members of P. falciparum were categorized into subfamilies. The ABC B subfamily was the largest and contained seven members. Other family members that could be involved in drug transport are PfABCC1, PfABCC2, PfABCG1, and PfABCI3. The expression and localization of three ABC transport proteins was determined. PfMRP1, PfMRP2, and PfMDR5 are localized to the plasma membrane in all asexual stages of the parasite. Conclusion In conclusion, 11 of the 16 ABC proteins in the P. falciparum genome are putative transport proteins, some of which might be involved in drug resistance. Moreover, it was demonstrated that three of these proteins are expressed on the parasite's plasma membrane.
Collapse
Affiliation(s)
- Reginald A Kavishe
- Department of Pharmacology and Toxicology 149, Radboud University Nijmegen Medical Centre, P,O, Box 9101, 6500 HB Nijmegen, the Netherlands.
| | | | | | | | | | | |
Collapse
|
45
|
Plasmodium falciparum drug resistance in Madagascar: facing the spread of unusual pfdhfr and pfmdr-1 haplotypes and the decrease of dihydroartemisinin susceptibility. Antimicrob Agents Chemother 2009; 53:4588-97. [PMID: 19704124 DOI: 10.1128/aac.00610-09] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The aim of this study was to provide the first comprehensive spatiotemporal picture of Plasmodium falciparum resistance in various geographic areas in Madagascar. Additional data about the antimalarial resistance in the neighboring islands of the Comoros archipelago were also collected. We assessed the prevalence of pfcrt, pfmdr-1, pfdhfr, and pfdhps mutations and the pfmdr-1 gene copy number in 1,596 P. falciparum isolates collected in 26 health centers (20 in Madagascar and 6 in the Comoros Islands) from 2006 to 2008. The in vitro responses to a panel of drugs by 373 of the parasite isolates were determined. The results showed (i) unusual profiles of chloroquine susceptibility in Madagascar, (ii) a rapid rise in the frequency of parasites with both the pfdhfr and the pfdhps mutations, (iii) the alarming emergence of the single pfdhfr 164L genotype, and (iv) the progressive loss of the most susceptible isolates to artemisinin derivatives. In the context of the implementation of the new national policy for the fight against malaria, continued surveillance for the detection of P. falciparum resistance in the future is required.
Collapse
|
46
|
Plasmodium falciparum Na+/H+ exchanger 1 transporter is involved in reduced susceptibility to quinine. Antimicrob Agents Chemother 2009; 53:1926-30. [PMID: 19273668 DOI: 10.1128/aac.01243-08] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Polymorphisms in the Plasmodium falciparum crt (Pfcrt), Pfmdr1, and Pfmrp genes were not significantly associated with quinine (QN) 50% inhibitory concentrations (IC(50)s) in 23 strains of Plasmodium falciparum. An increased number of DNNND repeats in Pfnhe-1 microsatellite ms4760 was associated with an increased IC(50) of QN (P = 0.0007). Strains with only one DNNND repeat were more susceptible to QN (mean IC(50) of 154 nM). Strains with two DNNND repeats had intermediate susceptibility to QN (mean IC(50) of 548 nM). Strains with three DNNND repeats had reduced susceptibility to QN (mean IC(50) of 764 nM). Increased numbers of NHNDNHNNDDD repeats were associated with a decreased IC(50) of QN (P = 0.0020). Strains with profile 7 for Pfnhe-1 ms4760 (ms4760-7) were significantly associated with reduced QN susceptibility (mean IC(50) of 764 nM). The determination of DNNND and NHNDNHNNDDD repeats in Pfnhe-1 ms4760 could be a good marker of QN resistance and provide an attractive surveillance method to monitor temporal trends in P. falciparum susceptibility to QN. The validity of the markers should be further supported by analyzing more isolates.
Collapse
|
47
|
Raj DK, Mu J, Jiang H, Kabat J, Singh S, Sullivan M, Fay MP, McCutchan TF, Su XZ. Disruption of a Plasmodium falciparum multidrug resistance-associated protein (PfMRP) alters its fitness and transport of antimalarial drugs and glutathione. J Biol Chem 2008; 284:7687-96. [PMID: 19117944 PMCID: PMC2658063 DOI: 10.1074/jbc.m806944200] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
ATP-binding cassette transporters play an important role in drug resistance
and nutrient transport. In the human malaria parasite Plasmodium
falciparum, a homolog of the human p-glycoprotein (PfPgh-1) was
shown to be involved in resistance to several drugs. More recently, many
transporters were associated with higher IC50 levels in responses
to chloroquine (CQ) and quinine (QN) in field isolates. Subsequent studies,
however, could not confirm the associations, although inaccuracy in drug tests
in the later studies could contribute to the lack of associations. Here we
disrupted a gene encoding a putative multidrug resistance-associated protein
(PfMRP) that was previously shown to be associated with P. falciparum
responses to CQ and QN. Parasites with disrupted PfMRP (W2/MRPΔ) could
not grow to a parasitemia higher than 5% under normal culture conditions,
possibly because of lower efficiency in removing toxic metabolites. The
W2/MRPΔ parasite also accumulated more radioactive glutathione, CQ, and
QN and became more sensitive to multiple antimalarial drugs, including CQ, QN,
artemisinin, piperaquine, and primaquine. PfMRP was localized on the parasite
surface membrane, within membrane-bound vesicles, and along the straight side
of the D-shaped stage II gametocytes. The results suggest that PfMRP plays a
role in the efflux of glutathione, CQ, and QN and contributes to parasite
responses to multiple antimalarial drugs, possibly by pumping drugs outside
the parasite.
Collapse
Affiliation(s)
- Dipak Kumar Raj
- Laboratory of Malaria and Vector Research, Research Technologies Branch, and Biostatistical Research Branch, NIAID, National Institutes of Health, Bethesda, Maryland 20892-8132, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
The human malaria parasite, Plasmodium falciparum, has long been known to have a homologue of the human 'multidrug resistance' P-glycoprotein. P-glycoprotein is an ABC transporter that pumps drugs from multidrug-resistant cancer cells. The malaria parasite's P-glycoprotein homologue, Pgh1, is known to influence the sensitivity of malaria parasites to a diverse range of antimalarial drugs, but the mechanism by which it does so has remained obscure. In a new paper, Sanchez et al. report the successful functional expression of Pgh1 in Xenopus laevis oocytes and provide the first direct demonstration of the ability of Pgh1 to transport drugs. The work provides important new insights into the mechanism by which Pgh1 influences malaria parasite drug sensitivity.
Collapse
Affiliation(s)
- Kevin J Saliba
- Biochemistry and Molecular Biology, School of Biology, The Australian National University, Canberra, ACT 0200, Australia.
| | | | | |
Collapse
|