151
|
Yao Y, Wu K, Xu M, Yang Y, Zhang Y, Yang W, Shang R, Du W, Tan H, Chen J, Lin M, Li J. Surveillance of Genetic Variations Associated with Antimalarial Resistance of Plasmodium falciparum Isolates from Returned Migrant Workers in Wuhan, Central China. Antimicrob Agents Chemother 2018; 62:AAC.02387-17. [PMID: 29941645 PMCID: PMC6125563 DOI: 10.1128/aac.02387-17] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 06/15/2018] [Indexed: 02/05/2023] Open
Abstract
Antimalarial drug resistance developed in Plasmodium falciparum has become a problem for malaria control. Evaluation of drug resistance is the first step for effective malaria control. In this study, we investigated the gene mutations of P. falciparum using blood samples from returned Chinese migrant workers in order to identify drug resistance-associated molecular markers. These workers returned from Africa and Southeast Asia (SEA) during 2011 to 2016. Polymorphisms in pfcrt, pfmdr1, and k13-propeller genes and the haplotype patterns of Pfcrt and Pfmdr1 were analyzed. The results showed the presence of four haplotypes of Pfcrt codons 72 to 76, including CVMNK (wild type), SVMNT and CVIET (mutation types), and CV M/I N/E K/T (mixed type), with 50.57%, 1.14%, 25.00%, and 23.30% prevalence, respectively. For Pfmdr1, N86Y (22.28%) and Y184F (60.01%) were the main prevalent mutations (mutations are underlined). The prevalence of mutation at position 550, 561, 575, and 589 of K13-propeller were 1.09%, 0.54%, 0.54%, and 0.54%, respectively. These data suggested that Pfcrt, Pfmdr1, and K13-propeller polymorphisms are potential markers to assess drug resistance of P. falciparum in China, Africa, and SEA.
Collapse
Affiliation(s)
- Yi Yao
- Department of Human Parasitology, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Department of Infectious Diseases, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Kai Wu
- Department of Schistosomiasis and Endemic Diseases, Wuhan City Center for Disease Prevention and Control, Wuhan, China
| | - Mingxing Xu
- Department of Schistosomiasis and Endemic Diseases, Wuhan City Center for Disease Prevention and Control, Wuhan, China
| | - Yan Yang
- Department of Schistosomiasis and Endemic Diseases, Wuhan City Center for Disease Prevention and Control, Wuhan, China
| | - Yijing Zhang
- Department of Human Parasitology, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Department of Infectious Diseases, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Wenjing Yang
- Department of Human Parasitology, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Department of Infectious Diseases, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Ronghua Shang
- Department of Human Parasitology, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Department of Infectious Diseases, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Weixing Du
- Department of Human Parasitology, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Department of Infectious Diseases, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Huabing Tan
- Department of Human Parasitology, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Department of Infectious Diseases, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Jiangtao Chen
- Laboratory Medical Center, Huizhou Municipal Central Hospital, Huizhou, China
| | - Min Lin
- Department of Histology and Embryology, Shantou University Medical College, Shantou, China
| | - Jian Li
- Department of Human Parasitology, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Department of Infectious Diseases, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| |
Collapse
|
152
|
Mechanisms of resistance to the partner drugs of artemisinin in the malaria parasite. Curr Opin Pharmacol 2018; 42:71-80. [PMID: 30142480 DOI: 10.1016/j.coph.2018.07.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 07/19/2018] [Accepted: 07/26/2018] [Indexed: 01/24/2023]
Abstract
The deployment of artemisinin-based combination therapies (ACTs) has been, and continues to be, integral to reducing the number of malaria cases and deaths. However, their efficacy is being increasingly jeopardized by the emergence and spread of parasites that are resistant (or partially resistant) to the artemisinin derivatives and to their partner drugs, with the efficacy of the latter being especially crucial for treatment success. A detailed understanding of the genetic determinants of resistance to the ACT partner drugs, and the mechanisms by which they mediate resistance, is required for the surveillance of molecular markers and to optimize the efficacy and lifespan of the partner drugs through resistance management strategies. We summarize new insights into the molecular basis of parasite resistance to the ACTs, such as recently-uncovered determinants of parasite susceptibility to the artemisinin derivatives, piperaquine, lumefantrine, and mefloquine, and outline the mechanisms through which polymorphisms in these determinants may be conferring resistance.
Collapse
|
153
|
Emerging Southeast Asian PfCRT mutations confer Plasmodium falciparum resistance to the first-line antimalarial piperaquine. Nat Commun 2018; 9:3314. [PMID: 30115924 PMCID: PMC6095916 DOI: 10.1038/s41467-018-05652-0] [Citation(s) in RCA: 165] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 07/11/2018] [Indexed: 11/16/2022] Open
Abstract
The widely used antimalarial combination therapy dihydroartemisinin + piperaquine (DHA + PPQ) has failed in Cambodia. Here, we perform a genomic analysis that reveals a rapid increase in the prevalence of novel mutations in the Plasmodium falciparum chloroquine resistance transporter PfCRT following DHA + PPQ implementation. These mutations occur in parasites harboring the K13 C580Y artemisinin resistance marker. By introducing PfCRT mutations into sensitive Dd2 parasites or removing them from resistant Cambodian isolates, we show that the H97Y, F145I, M343L, or G353V mutations each confer resistance to PPQ, albeit with fitness costs for all but M343L. These mutations sensitize Dd2 parasites to chloroquine, amodiaquine, and quinine. In Dd2 parasites, multicopy plasmepsin 2, a candidate molecular marker, is not necessary for PPQ resistance. Distended digestive vacuoles were observed in pfcrt-edited Dd2 parasites but not in Cambodian isolates. Our findings provide compelling evidence that emerging mutations in PfCRT can serve as a molecular marker and mediator of PPQ resistance. Increasing resistance of Plasmodium falciparum strains to piperaquine (PPQ) in Southeast Asia is of concern and resistance mechanisms are incompletely understood. Here, Ross et al. show that mutations in the P. falciparum chloroquine resistance transporter are rapidly increasing in prevalence in Cambodia and confer resistance to PPQ.
Collapse
|
154
|
Voumbo-Matoumona DF, Akiana J, Madamet M, Kouna LC, Lekana-Douki JB, Pradines B. High prevalence of Plasmodium falciparum antimalarial drug resistance markers in isolates from asymptomatic patients from the Republic of the Congo between 2010 and 2015. J Glob Antimicrob Resist 2018; 14:277-283. [PMID: 30121345 DOI: 10.1016/j.jgar.2018.08.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 08/01/2018] [Accepted: 08/02/2018] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVES This study investigated the prevalence of haplotypes of the Pfdhps, Pfdhfr, Pfcrt, Pfmdr1 and PfK13 resistance markers in isolates from asymptomatic patients from the Republic of the Congo following implementation of artemisinin based-combination therapy (ACT). METHODS Peripheral blood was collected from asymptomatic children in 2010 and 2015 from Brazzaville in the south and in 2013 in the north of the Congo. Genotypes of Pfmdr1, Pfcrt, Pfdhps, Pfdhfr and PfK13 were assessed by PCR. RESULTS Children from 2010 were younger than those from 2015 (mean age 5.38 years vs. 8.67 years; P=0.003). The main Pfcrt haplotype was the wild-type CVMNK (84.85%) in 2010, whereas the mutant CVIET (61.64%) predominated in 2015 (P<0.001). In the north, 45.00% of samples were CVMNK and 10.00% were CVIET. Other samples harboured new haplotypes in the country or mixed alleles. No significant difference in Pfmdr1 haplotypes was observed in 2010 and 2015 and the main haplotypes were NYD and NFD (30.56% vs. 28.57% and 61.11% vs. 42.86% for 2010 and 2015, respectively). In the south, the Pfdhps haplotypes observed were AAKAA, AGKAA, SGKAA and SGEGA (87.50% vs. 0%, 12.50% vs. 33.33%, 0% vs. 33.33% and 0% vs. 33.33% for 2010 and 2015, respectively). For Pfdhfr, the IRNI haplotype was most prevalent (85.71% for 2010, 87.50% for 2013 and 100% for 2015). No PfK13 mutations were found. CONCLUSIONS Monitoring the efficacy of ACT and intermittent preventive treatment with sulfadoxine-pyrimethamine is necessary to ensure an epidemiological survey of asymptomatic malaria.
Collapse
Affiliation(s)
- Dominique Fatima Voumbo-Matoumona
- Unité d'Evolution, Epidémiologie et Résistances Parasitaires (UNEEREP), Centre International de Recherche Médicales de Franceville, BP 769 Franceville, Gabon; Unité Parasitologie et Entomologie, Département des Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, Marseille, France; Ecole Doctorale Régionale d'Afrique Centrale en Infectiologie Tropicale, BP 876 Franceville, Gabon
| | - Jean Akiana
- Départements des Masters/Licences, Parcours-Types des Sciences Biologiques, Faculté des Sciences et Techniques, Université Marien Ngouabi, BP 69, Brazzaville, Congo; Direction de la Médecine Préventive et des Essais Cliniques, Laboratoire National de Santé Publique, BP 120 Brazzaville, Congo
| | - Marylin Madamet
- Unité Parasitologie et Entomologie, Département des Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, Marseille, France; Aix-Marseille Univ., IRD, AP-HM, SSA, VITROME, IHU Méditerranée Infection, Marseille, France; Centre National de Référence du Paludisme, Marseille, France
| | - Lady Charlène Kouna
- Unité d'Evolution, Epidémiologie et Résistances Parasitaires (UNEEREP), Centre International de Recherche Médicales de Franceville, BP 769 Franceville, Gabon
| | - Jean Bernard Lekana-Douki
- Unité d'Evolution, Epidémiologie et Résistances Parasitaires (UNEEREP), Centre International de Recherche Médicales de Franceville, BP 769 Franceville, Gabon; Département de Parasitologie Mycologie et de Médecine Tropicale, Université des Science de la Santé, BP 4005 Libreville, Gabon
| | - Bruno Pradines
- Unité Parasitologie et Entomologie, Département des Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, Marseille, France; Aix-Marseille Univ., IRD, AP-HM, SSA, VITROME, IHU Méditerranée Infection, Marseille, France; Centre National de Référence du Paludisme, Marseille, France.
| |
Collapse
|
155
|
Reiling SJ, Krohne G, Friedrich O, Geary TG, Rohrbach P. Chloroquine exposure triggers distinct cellular responses in sensitive versus resistant Plasmodium falciparum parasites. Sci Rep 2018; 8:11137. [PMID: 30042399 PMCID: PMC6057915 DOI: 10.1038/s41598-018-29422-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 07/06/2018] [Indexed: 11/25/2022] Open
Abstract
Chloroquine (CQ) treatment failure in Plasmodium falciparum parasites has been documented for decades, but the pharmacological explanation of this phenotype is not fully understood. Current concepts attribute CQ resistance to reduced accumulation of the drug at a given external CQ concentration ([CQ]ex) in resistant compared to sensitive parasites. The implication of this explanation is that the mechanisms of CQ-induced toxicity in resistant and sensitive strains are similar once lethal internal concentrations have been reached. To test this hypothesis, we investigated the mechanism of CQ-induced toxicity in CQ-sensitive (CQS) versus CQ-resistant (CQR) parasites by analyzing the time-course of cellular responses in these strains after exposure to varying [CQ]ex as determined in 72 h toxicity assays. Parasite killing was delayed in CQR parasites for up to 10 h compared to CQS parasites when exposed to equipotent [CQ]ex. In striking contrast, brief exposure (1 h) to lethal [CQ]ex in CQS but not CQR parasites caused the appearance of hitherto undescribed hemozoin (Hz)-containing compartments in the parasite cytosol. Hz-containing compartments were very rarely observed in CQR parasites even after CQ exposures sufficient to cause irreversible cell death. These findings challenge current concepts that CQ killing of malaria parasites is solely concentration-dependent, and instead suggest that CQS and CQR strains fundamentally differ in the consequences of CQ exposure.
Collapse
Affiliation(s)
- Sarah J Reiling
- Institute of Parasitology, McGill University, Ste. Anne de Bellevue (Montréal), Québec, Canada
| | - Georg Krohne
- Theodor Boveri Institute, Biocenter, University of Würzburg, Würzburg, Germany
| | - Oliver Friedrich
- Institute of Medical Biotechnology, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Timothy G Geary
- Institute of Parasitology, McGill University, Ste. Anne de Bellevue (Montréal), Québec, Canada
| | - Petra Rohrbach
- Institute of Parasitology, McGill University, Ste. Anne de Bellevue (Montréal), Québec, Canada.
| |
Collapse
|
156
|
Russo G, L'Episcopia M, Menegon M, Souza SS, Dongho BGD, Vullo V, Lucchi NW, Severini C. Dihydroartemisinin-piperaquine treatment failure in uncomplicated Plasmodium falciparum malaria case imported from Ethiopia. Infection 2018; 46:867-870. [PMID: 29980936 DOI: 10.1007/s15010-018-1174-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 07/04/2018] [Indexed: 12/15/2022]
Abstract
Dihydroartemisinin-piperaquine (DHA-PPQ) is the artemisinin combination therapy that was recently introduced for the treatment of Plasmodium falciparum uncomplicated malaria, but emerging resistance in South-East Asia is threatening its use. This report describes a case of DHA-PPQ treatment failure in uncomplicated malaria occurring in an immigrant living in Italy, after a travel to Ethiopia. Thirty days after malaria recovery following DHA-PPQ therapy, the patient had malaria recrudescence. According to the genotyping analysis, the same P. falciparum was responsible for both episodes. Thus, it seems important to consider possible malaria recrudescence occurring after DHA-PPQ therapy in patients from African countries.
Collapse
Affiliation(s)
- Gianluca Russo
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy.
| | - Mariangela L'Episcopia
- Department of Infectious Diseases, Istituto Superiore di Sanità, Viale Regina Elena Rome 299, 00161, Rome, Italy
| | - Michela Menegon
- Department of Infectious Diseases, Istituto Superiore di Sanità, Viale Regina Elena Rome 299, 00161, Rome, Italy
| | - Samaly Santos Souza
- Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention, Malaria Branch, 1600 Clifton Rd, Atlanta, GA, 30333, USA
| | | | - Vincenzo Vullo
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Naomi W Lucchi
- Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention, Malaria Branch, 1600 Clifton Rd, Atlanta, GA, 30333, USA
| | - Carlo Severini
- Department of Infectious Diseases, Istituto Superiore di Sanità, Viale Regina Elena Rome 299, 00161, Rome, Italy
| |
Collapse
|
157
|
Kittichai V, Nguitragool W, Ngassa Mbenda HG, Sattabongkot J, Cui L. Genetic diversity of the Plasmodium vivax multidrug resistance 1 gene in Thai parasite populations. INFECTION GENETICS AND EVOLUTION 2018; 64:168-177. [PMID: 29936038 DOI: 10.1016/j.meegid.2018.06.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 06/18/2018] [Accepted: 06/20/2018] [Indexed: 11/19/2022]
Abstract
Plasmodium vivax resistance to chloroquine (CQ) was first reported over 60 years ago. Here we analyzed sequence variations in the multidrug resistance 1 gene (Pvmdr1), a putative molecular marker for P. vivax CQ resistance, in field isolates collected from three sites in Thailand during 2013-2016. Several single nucleotide polymorphisms previously implicated in reduced CQ sensitivity were found. These genetic variations encode amino acids in the two nucleotide-binding domains as well as the transmembrane domains of the protein. The high level of genetic diversity of Pvmdr1 provides insights into the evolutionary history of this gene. Specifically, there was little evidence of positive selection at amino acid F1076L in global isolates to be promoted as a possible marker for CQ resistance. Population genetic analysis clearly divided the parasites into eastern and western populations, which is consistent with their geographical separation by the central malaria-free area of Thailand. With CQ-primaquine remaining as the frontline treatment for vivax malaria in all regions of Thailand, such a population subdivision could be shaped and affected by the current drugs for P. falciparum since mixed P. falciparum/P. vivax infections often occur in this region.
Collapse
Affiliation(s)
- Veerayuth Kittichai
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand; Faculty of Medicine, King Mongkut's Institute of Technology Ladkrabang, Bangkok, Thailand
| | - Wang Nguitragool
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | | | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand.
| | - Liwang Cui
- Department of Entomology, Center for Malaria Research, Pennsylvania State University, University Park, PA, USA.
| |
Collapse
|
158
|
Aguiar ACC, Panciera M, Simão dos Santos EF, Singh MK, Garcia ML, de Souza GE, Nakabashi M, Costa JL, Garcia CRS, Oliva G, Correia CRD, Guido RVC. Discovery of Marinoquinolines as Potent and Fast-Acting Plasmodium falciparum Inhibitors with in Vivo Activity. J Med Chem 2018; 61:5547-5568. [DOI: 10.1021/acs.jmedchem.8b00143] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- Anna Caroline Campos Aguiar
- Sao Carlos Institute of Physics, University of Sao Paulo, Av. Joao Dagnone, 1100 Jardim Santa Angelina, São Carlos, SP 13563-120, Brazil
| | - Michele Panciera
- Institute of Chemistry, State University of Campinas, Josue de Castro St., Campinas, SP 13083-970, Brazil
| | | | - Maneesh Kumar Singh
- Faculty of Pharmaceutical Sciences, University of Sao Paulo, Av. Prof. Lineu Prestes, 580 Cidade Universitária, São Paulo, SP 05508-900, Brazil
- Department of Physiology, University of Sao Paulo, Rua do Matão 101, Travessa 14, São Paulo, SP 05508-090, Brazil
| | - Mariana Lopes Garcia
- Sao Carlos Institute of Physics, University of Sao Paulo, Av. Joao Dagnone, 1100 Jardim Santa Angelina, São Carlos, SP 13563-120, Brazil
| | - Guilherme Eduardo de Souza
- Sao Carlos Institute of Physics, University of Sao Paulo, Av. Joao Dagnone, 1100 Jardim Santa Angelina, São Carlos, SP 13563-120, Brazil
| | - Myna Nakabashi
- Department of Physiology, University of Sao Paulo, Rua do Matão 101, Travessa 14, São Paulo, SP 05508-090, Brazil
| | - José Luiz Costa
- Faculty of Pharmaceutical Sciences, State University of Campinas, Rua Oswaldo Cruz, 2° Andar, Bloco F3, Cidade Universitária, Campinas, SP 13083-859, Brazil
| | - Célia R. S. Garcia
- Faculty of Pharmaceutical Sciences, University of Sao Paulo, Av. Prof. Lineu Prestes, 580 Cidade Universitária, São Paulo, SP 05508-900, Brazil
| | - Glaucius Oliva
- Sao Carlos Institute of Physics, University of Sao Paulo, Av. Joao Dagnone, 1100 Jardim Santa Angelina, São Carlos, SP 13563-120, Brazil
| | | | - Rafael Victorio Carvalho Guido
- Sao Carlos Institute of Physics, University of Sao Paulo, Av. Joao Dagnone, 1100 Jardim Santa Angelina, São Carlos, SP 13563-120, Brazil
| |
Collapse
|
159
|
Goomber S, Mishra N, Anvikar A, Yadav CP, Valecha N. Spatio-temporal distribution of PfMDR1 polymorphism among uncomplicated Plasmodium falciparum malaria cases along international border of north east India. INFECTION GENETICS AND EVOLUTION 2018; 63:285-290. [PMID: 29842979 DOI: 10.1016/j.meegid.2018.05.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 05/22/2018] [Accepted: 05/25/2018] [Indexed: 10/16/2022]
Abstract
PfMDR1 single nucleotide polymorphisms (SNP) are good correlate markers for antimalarial drug resistance worldwide. Present study is a comprehensive view of screening of PfMDR1 polymorphism to antimalarials practiced with geography and time. Study sites Mizoram, Tripura, Meghalaya chosen are at multivariate drug pressure due to cross border migration and transmission. Mizoram is gateway to south east Asia through Myanmar whereas Tripura, Meghalaya share porous border with Bangladesh. Baseline finger pricked blood stained filter paper for confirmed uncomplicated Plasmodium falciparum infected patients (year 2015) were obtained from National Institute of Malaria Research, New Delhi, India. PfMDR1 polymorphism for codon N86Y, Y184F, D1246Y was determined by PCR-RFLP, further confirmed by sequencing. There observed marked predominance of Plasmodium isolates with PfMDR1 wild type alleles for all codons under study i.e. 86, 184, 1246. Spatially, Plasmodium isolates from Mizoram were most diverse with co-existence of PfMDR1 genotype with NYD, YYD, NFD haplotypes, followed by Tripura. Isolates from Meghalaya were of all NYD haplotype. Reports, referring to screening of PfMDR1 SNPs to CQ/SP/AS-SP across India, were archived. Temporal study show distinct rise in proportion of PfMDR1 wild type N86 allele since introduction of Artemether-Lumefantrine as first line antimalarial. Hence spatio-temporal screening of Plasmodium population with PfMDR1 single nucleotide polymorphism accounts for its association with antimalarial susceptibility and validate PfMDR1 SNPs as antimalarial drug resistant marker.
Collapse
Affiliation(s)
- Shelly Goomber
- National Institute of Malaria Research, Indian Council of Medical Research, Sector - 8, Dwarka, New Delhi, India.
| | - Neelima Mishra
- National Institute of Malaria Research, Indian Council of Medical Research, Sector - 8, Dwarka, New Delhi, India
| | - Anup Anvikar
- National Institute of Malaria Research, Indian Council of Medical Research, Sector - 8, Dwarka, New Delhi, India
| | - Chander Prakash Yadav
- National Institute of Malaria Research, Indian Council of Medical Research, Sector - 8, Dwarka, New Delhi, India
| | - Neena Valecha
- National Institute of Malaria Research, Indian Council of Medical Research, Sector - 8, Dwarka, New Delhi, India
| |
Collapse
|
160
|
Identity-by-descent analyses for measuring population dynamics and selection in recombining pathogens. PLoS Genet 2018; 14:e1007279. [PMID: 29791438 PMCID: PMC5988311 DOI: 10.1371/journal.pgen.1007279] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 06/05/2018] [Accepted: 02/26/2018] [Indexed: 12/30/2022] Open
Abstract
Identification of genomic regions that are identical by descent (IBD) has proven useful for human genetic studies where analyses have led to the discovery of familial relatedness and fine-mapping of disease critical regions. Unfortunately however, IBD analyses have been underutilized in analysis of other organisms, including human pathogens. This is in part due to the lack of statistical methodologies for non-diploid genomes in addition to the added complexity of multiclonal infections. As such, we have developed an IBD methodology, called isoRelate, for analysis of haploid recombining microorganisms in the presence of multiclonal infections. Using the inferred IBD status at genomic locations, we have also developed a novel statistic for identifying loci under positive selection and propose relatedness networks as a means of exploring shared haplotypes within populations. We evaluate the performance of our methodologies for detecting IBD and selection, including comparisons with existing tools, then perform an exploratory analysis of whole genome sequencing data from a global Plasmodium falciparum dataset of more than 2500 genomes. This analysis identifies Southeast Asia as having many highly related isolates, possibly as a result of both reduced transmission from intensified control efforts and population bottlenecks following the emergence of antimalarial drug resistance. Many signals of selection are also identified, most of which overlap genes that are known to be associated with drug resistance, in addition to two novel signals observed in multiple countries that have yet to be explored in detail. Additionally, we investigate relatedness networks over the selected loci and determine that one of these sweeps has spread between continents while the other has arisen independently in different countries. IBD analysis of microorganisms using isoRelate can be used for exploring population structure, positive selection and haplotype distributions, and will be a valuable tool for monitoring disease control and elimination efforts of many diseases. There are growing concerns over the emergence of antimicrobial drug resistance, which threatens the efficacy of treatments for infectious diseases such as malaria. As such, it is important to understand the dynamics of resistance by investigating population structure, natural selection and disease transmission in microorganisms. The study of disease dynamics has been hampered by the lack of suitable statistical models for analysis of isolates containing multiple infections. We introduce a statistical model that uses population genomic data to identify genomic regions (loci) that are inherited from a common ancestor, in the presence of multiple infections. We demonstrate its potential for biological discovery using a global Plasmodium falciparum dataset. We identify low genetic diversity in isolates from Southeast Asia, possibly from clonal expansion following intensified control efforts after the emergence of artemisinin resistance. We also identify loci under positive selection, most of which contain genes that have been associated with antimalarial drug resistance. We discover two loci under strong selection in multiple countries throughout Southeast Asia and Africa where the selection pressure is currently unknown. We find that the selection pressure at one of these loci has originated from gene flow, while the other loci has originated from multiple independent events.
Collapse
|
161
|
Thita T, Jadsri P, Thamkhantho J, Ruang-Areerate T, Suwandittakul N, Sitthichot N, Mahotorn K, Tan-Ariya P, Mungthin M. Phenotypic and genotypic characterization of Thai isolates of Plasmodium falciparum after an artemisinin resistance containment project. Malar J 2018; 17:197. [PMID: 29764451 PMCID: PMC5952644 DOI: 10.1186/s12936-018-2347-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 05/07/2018] [Indexed: 11/28/2022] Open
Abstract
Background In Thailand, artemisinin-based combination therapy (ACT) has been used to treat uncomplicated falciparum malaria since 1995. Unfortunately, artemisinin resistance has been reported from Thailand and other Southeast Asian countries since 2003. Malarone®, a combination of atovaquone–proguanil (ATQ–PG), has been used to cease artemisinin pressure in some areas along Thai–Cambodia border, as part of an artemisinin resistance containment project since 2009. This study aimed to determine genotypes and phenotypes of Plasmodium falciparum isolates collected from the Thai–Cambodia border after the artemisinin resistance containment project compared with those collected before. Results One hundred and nine of P. falciparum isolates collected from Thai–Cambodia border from Chanthaburi and Trat provinces during 1988–2016 were used in this study. Of these, 58 isolates were collected after the containment. These parasite isolates were characterized for in vitro antimalarial sensitivities including chloroquine (CQ), quinine (QN), mefloquine (MQ), piperaquine (PPQ), artesunate (AS), dihydroartemisinin (DHA), ATQ and PG and genetic markers for drug resistance including the Kelch13 (k13), Plasmodium falciparum chloroquine resistance transporter (pfcrt), P. falciparum multidrug resistance 1 (pfmdr1) and cytochrome b (cytb) genes. Mean CQ, QN, MQ, PPQ and AS IC50s of the parasite isolates collected from 2009 to 2016 exhibited significantly higher than those of parasites collected before 2009. Approximately 57% exhibited in vitro MQ resistance. Approximately 94% of the isolates collected from 2009 to 2016 contained the pfmdr1 184F allele. Mutations of the k13 gene were detected in approximately 90% of the parasites collected from 2009 to 2016 which were significantly higher than the parasite isolates collected before. No ATQ-resistant genotype and phenotype of P. falciparum were found among the isolates collected after the containment project. Conclusions Although the containment project had been implemented in this area, the expansion of artemisinin-resistant parasites did not decline. In addition, reduced sensitivity of the partner drugs of ACT including MQ and PPQ were identified.
Collapse
Affiliation(s)
- Thunyapit Thita
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Pimrat Jadsri
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Jarupatr Thamkhantho
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Toon Ruang-Areerate
- Department of Parasitology, Phramongkutklao College of Medicine, Bangkok, Thailand
| | | | - Naruemon Sitthichot
- Department of Parasitology, Phramongkutklao College of Medicine, Bangkok, Thailand
| | - Kittiya Mahotorn
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Peerapan Tan-Ariya
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Mathirut Mungthin
- Department of Parasitology, Phramongkutklao College of Medicine, Bangkok, Thailand.
| |
Collapse
|
162
|
Next-Generation Sequencing and Bioinformatics Protocol for Malaria Drug Resistance Marker Surveillance. Antimicrob Agents Chemother 2018; 62:AAC.02474-17. [PMID: 29439965 DOI: 10.1128/aac.02474-17] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 01/29/2018] [Indexed: 11/20/2022] Open
Abstract
The recent advances in next-generation sequencing technologies provide a new and effective way of tracking malaria drug-resistant parasites. To take advantage of this technology, an end-to-end Illumina targeted amplicon deep sequencing (TADS) and bioinformatics pipeline for molecular surveillance of drug resistance in P. falciparum, called malaria resistance surveillance (MaRS), was developed. TADS relies on PCR enriching genomic regions, specifically target genes of interest, prior to deep sequencing. MaRS enables researchers to simultaneously collect data on allele frequencies of multiple full-length P. falciparum drug resistance genes (crt, mdr1, k13, dhfr, dhps, and the cytochrome b gene), as well as the mitochondrial genome. Information is captured at the individual patient level for both known and potential new single nucleotide polymorphisms associated with drug resistance. The MaRS pipeline was validated using 245 imported malaria cases that were reported to the Centers for Disease Control and Prevention (CDC). The chloroquine resistance crt CVIET genotype (mutations underlined) was observed in 42% of samples, the highly pyrimethamine-resistant dhpsIRN triple mutant in 92% of samples, and the sulfadoxine resistance dhps mutation SGEAA in 26% of samples. The mdr1 NFSND genotype was found in 40% of samples. With the exception of two cases imported from Cambodia, no artemisinin resistance k13 alleles were identified, and 99% of patients carried parasites susceptible to atovaquone-proguanil. Our goal is to implement MaRS at the CDC for routine surveillance of imported malaria cases in the United States and to aid in the adoption of this system at participating state public health laboratories, as well as by global partners.
Collapse
|
163
|
Abstract
Although the application of CRISPR/Cas9 genome engineering approaches was first reported in apicomplexan parasites only 3 years ago, this technology has rapidly become an essential component of research on apicomplexan parasites. This review briefly describes the history of CRISPR/Cas9 and the principles behind its use along with documenting its implementation in apicomplexan parasites, especially Plasmodium spp. and Toxoplasma gondii. We also discuss the recent use of CRISPR/Cas9 for whole genome screening of gene knockout mutants in T. gondii and highlight its use for seminal genetic manipulations of Cryptosporidium spp. Finally, we consider new variations of CRISPR/Cas9 that have yet to be implemented in apicomplexans. Whereas CRISPR/Cas9 has already accelerated rapid interrogation of gene function in apicomplexans, the full potential of this technology is yet to be realized as new variations and innovations are integrated into the field.
Collapse
|
164
|
Zhang T, Xu X, Jiang J, Yu C, Tian C, Li W. Surveillance of Antimalarial Resistance Molecular Markers in Imported Plasmodium falciparum Malaria Cases in Anhui, China, 2012-2016. Am J Trop Med Hyg 2018; 98:1132-1136. [PMID: 29436339 DOI: 10.4269/ajtmh.17-0864] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Between 2012 and 2016, over 80% of registered malaria cases in Anhui province were Plasmodium falciparum returned from Africa. However, drug-resistance marker polymorphisms in imported P. falciparum cases have not been assessed. This study looked at the distribution of antimalarial-drug resistance by evaluating K13-propeller, pfmdr1, and pfcrt gene mutations. Fourteen synonymous and 15 nonsynonymous mutations in the K13-propeller gene were detected in samples from nine African countries, yet no candidate and validated K13 resistance mutations were found. The prevalence of pfcrt K76T and pfmdr1 N86Y mutants was 27.7% and 19.9%, respectively. Six different pfcrt genotypes were found, with C72V73M74N75T76 being the most common (89.2%). The pfcrt 76-pfmdr1 86 haplotype combination was evaluated in 173 isolates, and the N86T76 genotype was the most prevalent (50.3%). Notably, the prevalence of the N86Y mutation in Africa marked a decline from 31.0% in 2012 to 8.2% in 2016. Our findings suggest that there is no immediate threat to artemisinin efficacy in imported P. falciparum infections returned from Africa to Anhui province. Nevertheless, pfcrt K76T and pfmdr1 N86Y mutations were modestly prevalent, suggesting the presence of chloroquine resistance in these cases. Accordingly, dihydroartemisinin + piperaquine may be a better choice than artesunate + amodiaquine for the treatment of uncomplicated P. falciparum infections in Anhui province. In addition to, artemether-lumefantrine can be introduced as an alternative measure.
Collapse
Affiliation(s)
- Tao Zhang
- Anhui Provincial Center for Disease Control and Prevention, Anhui, China
| | - Xian Xu
- Anhui Provincial Center for Disease Control and Prevention, Anhui, China
| | - Jingjing Jiang
- Anhui Provincial Center for Disease Control and Prevention, Anhui, China
| | - Chen Yu
- Anhui Provincial Center for Disease Control and Prevention, Anhui, China
| | - Cuicui Tian
- Anhui Provincial Center for Disease Control and Prevention, Anhui, China
| | - Weidong Li
- Anhui Provincial Center for Disease Control and Prevention, Anhui, China
| |
Collapse
|
165
|
Baraka V, Mavoko HM, Nabasumba C, Francis F, Lutumba P, Alifrangis M, Van geertruyden JP. Impact of treatment and re-treatment with artemether-lumefantrine and artesunate-amodiaquine on selection of Plasmodium falciparum multidrug resistance gene-1 polymorphisms in the Democratic Republic of Congo and Uganda. PLoS One 2018; 13:e0191922. [PMID: 29390014 PMCID: PMC5794077 DOI: 10.1371/journal.pone.0191922] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 01/10/2018] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND The emergence of resistance against artemisinin combination treatment is a major concern for malaria control. ACTs are recommended as the rescue treatment, however, there is limited evidence as to whether treatment and re-treatment with ACTs select for drug-resistant P. falciparum parasites. Thus, the purpose of the present study is to investigate the impact of (re-)treatment using artesunate-amodiaquine (ASAQ) and artemether-lumefantrine (AL) on the selection of P. falciparum multidrug resistance-1 (Pfmdr1) alleles in clinical settings. METHODS P. falciparum positive samples were collected from children aged 12-59 months in a clinical trial in DR Congo and Uganda. Pfmdr1 single nucleotide polymorphisms (SNPs) analysis at codons N86Y, Y184F, and D1246Y were performed at baseline and post-treatment with either AL or ASAQ as a rescue treatment using nested PCR followed by restriction fragment length polymorphism (RFLP) assays. RESULTS The pre-treatment prevalence of Pfmdr1 N86 and D1246Y varied significantly between the sites, (p>0.001) and (p = 0.013), respectively. There was borderline significant directional selection for Pfmdr1 184F in recurrent malaria infections after treatment with AL in Uganda site (p = 0.05). Pfmdr1 NFD haplotype did not significantly change in post-treatment infections after re-treatment with either AL or ASAQ. Comparison between pre-treatment and post-treatment recurrences did not indicate directional selection of Pfmdr1 N86, D1246 alleles in the pre-RCT, RCT and post-RCT phases in both AL and ASAQ treatment arms. Pfmdr1 86Y was significantly associated with reduced risk of AL treatment failure (RR = 0.34, 95% CI:0.11-1.05, p = 0.04) while no evidence for D1246 allele (RR = 1.02; 95% CI: 0.42-2.47, p = 1.0). Survival estimates showed that the Pfmdr1 alleles had comparable mean-time to PCR-corrected recrudescence and new infections in both AL and ASAQ treatment arms. CONCLUSION We found limited impact of (re-)treatment with AL or ASAQ on selection for Pfmdr1 variants and haplotypes associated with resistance to partner drugs. These findings further supplement the evidence use of same or alternative ACTs as a rescue therapy for recurrent P.falciparum infections. Continued monitoring of genetic signatures of resistance is warranted to timely inform malaria (re-)treatment policies and guidelines.
Collapse
Affiliation(s)
- Vito Baraka
- National Institute for Medical Research, Tanga Centre, Tanga, United Republic of Tanzania
- Global Health Institute, University of Antwerp, Antwerp, Belgium
| | - Hypolite Muhindo Mavoko
- Global Health Institute, University of Antwerp, Antwerp, Belgium
- Département de Médecine Tropicale, Faculté de Médecine, Université de Kinshasa, Kinshasa, Democratic Republic of Congo
| | - Carolyn Nabasumba
- Global Health Institute, University of Antwerp, Antwerp, Belgium
- Epicentre Mbarara Research Base, Mbarara, Uganda
| | - Filbert Francis
- National Institute for Medical Research, Tanga Centre, Tanga, United Republic of Tanzania
| | - Pascal Lutumba
- Département de Médecine Tropicale, Faculté de Médecine, Université de Kinshasa, Kinshasa, Democratic Republic of Congo
| | - Michael Alifrangis
- Centre for Medical Parasitology at the Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, National University Hospital (Rigshospitalet), Copenhagen, Denmark
| | | |
Collapse
|
166
|
Abstract
A marked decrease in malaria-related deaths worldwide has been attributed to the administration of effective antimalarials against Plasmodium falciparum, in particular, artemisinin-based combination therapies (ACTs). Increasingly, ACTs are also used to treat Plasmodium vivax, the second major human malaria parasite. However, resistance to frontline artemisinins and partner drugs is now causing the failure of P. falciparum ACTs in southeast Asia. In this Review, we discuss our current knowledge of markers and mechanisms of resistance to artemisinins and ACTs. In particular, we describe the identification of mutations in the propeller domains of Kelch 13 as the primary marker for artemisinin resistance in P. falciparum and explore two major mechanisms of resistance that have been independently proposed: the activation of the unfolded protein response and proteostatic dysregulation of parasite phosphatidylinositol 3- kinase. We emphasize the continuing challenges and the imminent need to understand mechanisms of resistance to improve parasite detection strategies, develop new combinations to eliminate resistant parasites and prevent their global spread.
Collapse
|
167
|
Disrupting CD147-RAP2 interaction abrogates erythrocyte invasion by Plasmodium falciparum. Blood 2018; 131:1111-1121. [PMID: 29352039 DOI: 10.1182/blood-2017-08-802918] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 01/08/2018] [Indexed: 11/20/2022] Open
Abstract
Effective vaccines against malaria caused by Plasmodium falciparum are still lacking, and the molecular mechanism of the host-parasite interaction is not fully understood. Here we demonstrate that the interaction of RAP2, a parasite-secreted rhoptry protein that functions in the parasitophorous vacuole formation stage of the invasion, and CD147 on the host erythrocyte is essential for erythrocyte invasion by P falciparum and is independent from all previously identified interactions involved. Importantly, the blockade of the CD147-RAP2 interaction by HP6H8, a humanized CD147 antibody, completely abolished the parasite invasion with both cure and preventative functions in a humanized mouse model. Together with its long half-life on human red blood cells and its safety profile in cynomolgus monkeys, HP6H8 is the first antibody that offers an advantageous approach by targeting a more conserved late-stage parasite ligand for preventing as well as treating severe malaria.
Collapse
|
168
|
Conrad MD, Mota D, Foster M, Tukwasibwe S, Legac J, Tumwebaze P, Whalen M, Kakuru A, Nayebare P, Wallender E, Havlir DV, Jagannathan P, Huang L, Aweeka F, Kamya MR, Dorsey G, Rosenthal PJ. Impact of Intermittent Preventive Treatment During Pregnancy on Plasmodium falciparum Drug Resistance-Mediating Polymorphisms in Uganda. J Infect Dis 2017; 216:1008-1017. [PMID: 28968782 PMCID: PMC5853776 DOI: 10.1093/infdis/jix421] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 08/17/2017] [Indexed: 11/13/2022] Open
Abstract
Background In a recent trial of intermittent preventive treatment in pregnancy (IPTp) in Uganda, dihydroartemisinin-piperaquine (DP) was superior to sulfadoxine-pyrimethamine (SP) in preventing maternal and placental malaria. Methods We compared genotypes using sequencing, fluorescent microsphere, and quantitative polymerase chain reaction assays at loci associated with drug resistance in Plasmodium falciparum isolated from subjects receiving DP or SP. Results Considering aminoquinoline resistance, DP was associated with increased prevalences of mutations at pfmdr1 N86Y, pfmdr1 Y184F, and pfcrt K76T compared to SP (64.6% vs 27.4%, P < .001; 93.9% vs 59.2%, P < .001; and 87.7% vs 75.4%, P = .03, respectively). Increasing plasma piperaquine concentration at the time of parasitemia was associated with increasing pfmdr1 86Y prevalence; no infections with the N86 genotype occurred with piperaquine >2.75 ng/mL. pfkelch13 propeller domain polymorphisms previously associated with artemisinin resistance were not identified. Recently identified markers of piperaquine resistance were uncommon and not associated with DP. Considering antifolate resistance, SP was associated with increased prevalence of a 5-mutation haplotype (pfdhfr 51I, 59R, and 108N; pfdhps 437G and 581G) compared to DP (90.8% vs 60.0%, P = .001). Conclusions IPTp selected for genotypes associated with decreased sensitivity to treatment regimens, but genotypes associated with clinically relevant DP resistance in Asia have not emerged in Uganda.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Abel Kakuru
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | | | | | | | - Prasanna Jagannathan
- University of California, San Francisco,Stanford University, Palo Alto, California
| | | | | | - Moses R Kamya
- Infectious Diseases Research Collaboration, Kampala, Uganda,Makerere University College of Health Sciences, Kampala, Uganda
| | | | - Philip J Rosenthal
- University of California, San Francisco,Correspondence: P. J. Rosenthal, University of California, San Francisco, Box 0811, San Francisco, CA 94143 ()
| |
Collapse
|
169
|
Songsungthong W, Kulawonganunchai S, Wilantho A, Tongsima S, Koonyosying P, Uthaipibull C, Kamchonwongpaisan S, Shaw PJ. The Plasmodium berghei RC strain is highly diverged and harbors putatively novel drug resistance variants. PeerJ 2017; 5:e3766. [PMID: 29018598 PMCID: PMC5632537 DOI: 10.7717/peerj.3766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 08/15/2017] [Indexed: 11/20/2022] Open
Abstract
Background The current first line drugs for treating uncomplicated malaria are artemisinin (ART) combination therapies. However, Plasmodium falciparum parasites resistant to ART and partner drugs are spreading, which threatens malaria control efforts. Rodent malaria species are useful models for understanding antimalarial resistance, in particular genetic variants responsible for cross resistance to different compounds. Methods The Plasmodium berghei RC strain (PbRC) is described as resistant to different antimalarials, including chloroquine (CQ) and ART. In an attempt to identify the genetic basis for the antimalarial resistance trait in PbRC, its genome was sequenced and compared with five other previously sequenced P. berghei strains. Results We found that PbRC is eight-fold less sensitive to the ART derivative artesunate than the reference strain PbANKA. The genome of PbRC is markedly different from other strains, and 6,974 single nucleotide variants private to PbRC were identified. Among these PbRC private variants, non-synonymous changes were identified in genes known to modulate antimalarial sensitivity in rodent malaria species, including notably the ubiquitin carboxyl-terminal hydrolase 1 gene. However, no variants were found in some genes with strong evidence of association with ART resistance in P. falciparum such as K13 propeller protein. Discussion The variants identified in PbRC provide insight into P. berghei genome diversity and genetic factors that could modulate CQ and ART resistance in Plasmodium spp.
Collapse
Affiliation(s)
- Warangkhana Songsungthong
- Protein-Ligand Engineering and Molecular Biology Laboratory, Medical Molecular Biology Research Unit, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani, Thailand.,Biomolecular Analysis and Application Laboratory, Biosensing Technology Research Unit, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani, Thailand
| | - Supasak Kulawonganunchai
- Biostatistics and Bioinformatics Laboratory, Genome Technology Research Unit, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani, Thailand
| | - Alisa Wilantho
- Biostatistics and Bioinformatics Laboratory, Genome Technology Research Unit, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani, Thailand
| | - Sissades Tongsima
- Biostatistics and Bioinformatics Laboratory, Genome Technology Research Unit, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani, Thailand
| | - Pongpisid Koonyosying
- Protein-Ligand Engineering and Molecular Biology Laboratory, Medical Molecular Biology Research Unit, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani, Thailand
| | - Chairat Uthaipibull
- Protein-Ligand Engineering and Molecular Biology Laboratory, Medical Molecular Biology Research Unit, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani, Thailand
| | - Sumalee Kamchonwongpaisan
- Protein-Ligand Engineering and Molecular Biology Laboratory, Medical Molecular Biology Research Unit, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani, Thailand
| | - Philip J Shaw
- Protein-Ligand Engineering and Molecular Biology Laboratory, Medical Molecular Biology Research Unit, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani, Thailand
| |
Collapse
|
170
|
Genomic analysis of Isometamidium Chloride resistance in Trypanosoma congolense. INTERNATIONAL JOURNAL FOR PARASITOLOGY-DRUGS AND DRUG RESISTANCE 2017; 7:350-361. [PMID: 29032180 PMCID: PMC5645165 DOI: 10.1016/j.ijpddr.2017.10.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 10/02/2017] [Accepted: 10/05/2017] [Indexed: 12/30/2022]
Abstract
Isometamidium Chloride (ISM) is one of the principal drugs used to counteract Trypanosoma congolense infection in livestock, both as a prophylactic as well as a curative treatment. However, numerous cases of ISM resistance have been reported in different African regions, representing a significant constraint in the battle against Animal African Trypanosomiasis. In order to identify genetic signatures associated with ISM resistance in T. congolense, the sensitive strain MSOROM7 was selected for induction of ISM resistance in a murine host. Administered ISM concentrations in immune-suppressed mice were gradually increased from 0.001 mg/kg to 1 mg/kg, the maximal dose used in livestock. As a result, three independent MSOROM7 lines acquired full resistance to this concentration after five months of induction, and retained this full resistant phenotype following a six months period without drug pressure. In contrast, parasites did not acquire ISM resistance in immune-competent animals, even after more than two years under ISM pressure, suggesting that the development of full ISM resistance is strongly enhanced when the host immune response is compromised. Genomic analyses comparing the ISM resistant lines with the parental sensitive line identified shifts in read depth at heterozygous loci in genes coding for different transporters and transmembrane products, and several of these shifts were also found within natural ISM resistant isolates. These findings suggested that the transport and accumulation of ISM inside the resistant parasites may be modified, which was confirmed by flow cytometry and ex vivo ISM uptake assays that showed a decrease in the accumulation of ISM in the resistant parasites.
Collapse
|
171
|
Blasco B, Leroy D, Fidock DA. Antimalarial drug resistance: linking Plasmodium falciparum parasite biology to the clinic. Nat Med 2017; 23:917-928. [PMID: 28777791 DOI: 10.1038/nm.4381] [Citation(s) in RCA: 327] [Impact Index Per Article: 46.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Accepted: 06/30/2017] [Indexed: 02/08/2023]
Abstract
The global adoption of artemisinin-based combination therapies (ACTs) in the early 2000s heralded a new era in effectively treating drug-resistant Plasmodium falciparum malaria. However, several Southeast Asian countries have now reported the emergence of parasites that have decreased susceptibility to artemisinin (ART) derivatives and ACT partner drugs, resulting in increasing rates of treatment failures. Here we review recent advances in understanding how antimalarials act and how resistance develops, and discuss new strategies for effectively combatting resistance, optimizing treatment and advancing the global campaign to eliminate malaria.
Collapse
Affiliation(s)
| | - Didier Leroy
- Medicines for Malaria Venture, Geneva, Switzerland
| | - David A Fidock
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, New York, USA.,Division of Infectious Diseases, Department of Medicine, Columbia University Medical Center, New York, New York, USA
| |
Collapse
|
172
|
Vanaerschot M, Lucantoni L, Li T, Combrinck JM, Ruecker A, Kumar TRS, Rubiano K, Ferreira PE, Siciliano G, Gulati S, Henrich PP, Ng CL, Murithi JM, Corey VC, Duffy S, Lieberman OJ, Veiga MI, Sinden RE, Alano P, Delves MJ, Lee Sim K, Winzeler EA, Egan TJ, Hoffman SL, Avery VM, Fidock DA. Hexahydroquinolines are antimalarial candidates with potent blood-stage and transmission-blocking activity. Nat Microbiol 2017. [PMID: 28808258 DOI: 10.1038/s41564-017-0007–4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Antimalarial compounds with dual therapeutic and transmission-blocking activity are desired as high-value partners for combination therapies. Here, we report the identification and characterization of hexahydroquinolines (HHQs) that show low nanomolar potency against both pathogenic and transmissible intra-erythrocytic forms of the malaria parasite Plasmodium falciparum. This activity translates into potent transmission-blocking potential, as shown by in vitro male gamete formation assays and reduced oocyst infection and prevalence in Anopheles mosquitoes. In vivo studies illustrated the ability of lead HHQs to suppress Plasmodium berghei blood-stage parasite proliferation. Resistance selection studies, confirmed by CRISPR-Cas9-based gene editing, identified the digestive vacuole membrane-spanning transporter PfMDR1 (P. falciparum multidrug resistance gene-1) as a determinant of parasite resistance to HHQs. Haemoglobin and haem fractionation assays suggest a mode of action that results in reduced haemozoin levels and might involve inhibition of host haemoglobin uptake into intra-erythrocytic parasites. Furthermore, parasites resistant to HHQs displayed increased susceptibility to several first-line antimalarial drugs, including lumefantrine, confirming that HHQs have a different mode of action to other antimalarials drugs for which PfMDR1 is known to confer resistance. This work evokes therapeutic strategies that combine opposing selective pressures on this parasite transporter as an approach to countering the emergence and transmission of multidrug-resistant P. falciparum malaria.
Collapse
Affiliation(s)
- Manu Vanaerschot
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Leonardo Lucantoni
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, 4111, Queensland, Australia
| | - Tao Li
- Sanaria Inc., Rockville, MD, 20852, USA
| | - Jill M Combrinck
- Division of Pharmacology, Department of Medicine, University of Cape Town, Cape Town, 7925, South Africa
| | - Andrea Ruecker
- Department of Life Sciences, Imperial College, London, SW7 2AZ, UK.,Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK
| | - T R Santha Kumar
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Kelly Rubiano
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Pedro E Ferreira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal
| | - Giulia Siciliano
- Dipartimento di Malattie Infettive, Parassitarie ed Immunomediate, Istituto Superiore di Sanità, 00161, Rome, Italy
| | - Sonia Gulati
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Philipp P Henrich
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Caroline L Ng
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY, 10032, USA
| | - James M Murithi
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Victoria C Corey
- University of California, San Diego, School of Medicine, La Jolla, CA, 92093, USA
| | - Sandra Duffy
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, 4111, Queensland, Australia
| | - Ori J Lieberman
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY, 10032, USA
| | - M Isabel Veiga
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal
| | - Robert E Sinden
- Department of Life Sciences, Imperial College, London, SW7 2AZ, UK
| | - Pietro Alano
- Dipartimento di Malattie Infettive, Parassitarie ed Immunomediate, Istituto Superiore di Sanità, 00161, Rome, Italy
| | - Michael J Delves
- Department of Life Sciences, Imperial College, London, SW7 2AZ, UK
| | | | - Elizabeth A Winzeler
- University of California, San Diego, School of Medicine, La Jolla, CA, 92093, USA
| | - Timothy J Egan
- Department of Chemistry, University of Cape Town, Cape Town, 7700, South Africa
| | | | - Vicky M Avery
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, 4111, Queensland, Australia
| | - 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
|
173
|
Saksena R, Matlani M, Singh V, Kumar A, Anveshi A, Kumar D, Gaind R. Early treatment failure in concurrent dengue and mixed malaria species infection with suspected resistance to artemisinin combination therapy from a tertiary care center in Delhi: a case report. Int Med Case Rep J 2017; 10:289-294. [PMID: 28860870 PMCID: PMC5566360 DOI: 10.2147/imcrj.s139729] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background Concurrent dengue and mixed malaria infections in a single patient present with overlapping clinical manifestations which pose a diagnostic challenge and management dilemma in areas of common endemicities. Methods We report a case of a young male who tested positive for both Plasmodium vivax and Plasmodium falciparum along with dengue infection. He showed signs of early treatment failure to artemisinin combination therapy (artesunate with sulfadoxine+pyrimethamine). Molecular analysis for the drug resistance genes viz: chloroquine resistance (pfcrt), multidrug resistance (pfmdr-1), sulfadoxine (pfdhps), pyrimethamine (pfdhfr), and artemisinin resistance (keltch 13) was performed. Results A rise in parasitemia from <2% to 5% was observed after 3 days of treatment. Mutations in pfcrt, pfmdr-1, pfdhfr, and pfdhps genes were detected as a possible cause of treatment failure. Conclusion Increased severity, overlapping symptoms, and suspected resistance to treatment warrants a multidimensional diagnostic approach and diligent therapeutic monitoring.
Collapse
Affiliation(s)
- Rushika Saksena
- Department of Microbiology, Vardhman Mahavir Medical College and Safdarjung Hospital, Delhi, India
| | - Monika Matlani
- Department of Microbiology, Vardhman Mahavir Medical College and Safdarjung Hospital, Delhi, India
| | - Vineeta Singh
- Cell Biology Laboratory and Malaria Parasite Bank, National Institute of Malaria Research, Delhi, India
| | - Amit Kumar
- Cell Biology Laboratory and Malaria Parasite Bank, National Institute of Malaria Research, Delhi, India
| | - Anupam Anveshi
- Department of Microbiology, Vardhman Mahavir Medical College and Safdarjung Hospital, Delhi, India
| | - Dilip Kumar
- Department of Internal Medicine, Vardhman Mahavir Medical College and Safdarjung Hospital, Delhi, India
| | - Rajni Gaind
- Department of Microbiology, Vardhman Mahavir Medical College and Safdarjung Hospital, Delhi, India
| |
Collapse
|
174
|
Vanaerschot M, Lucantoni L, Li T, Combrinck JM, Ruecker A, Kumar TRS, Rubiano K, Ferreira PE, Siciliano G, Gulati S, Henrich PP, Ng CL, Murithi JM, Corey VC, Duffy S, Lieberman OJ, Veiga MI, Sinden RE, Alano P, Delves MJ, Lee Sim K, Winzeler EA, Egan TJ, Hoffman SL, Avery VM, Fidock DA. Hexahydroquinolines are antimalarial candidates with potent blood-stage and transmission-blocking activity. Nat Microbiol 2017; 2:1403-1414. [PMID: 28808258 DOI: 10.1038/s41564-017-0007-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 07/11/2017] [Indexed: 12/21/2022]
Abstract
Antimalarial compounds with dual therapeutic and transmission-blocking activity are desired as high-value partners for combination therapies. Here, we report the identification and characterization of hexahydroquinolines (HHQs) that show low nanomolar potency against both pathogenic and transmissible intra-erythrocytic forms of the malaria parasite Plasmodium falciparum. This activity translates into potent transmission-blocking potential, as shown by in vitro male gamete formation assays and reduced oocyst infection and prevalence in Anopheles mosquitoes. In vivo studies illustrated the ability of lead HHQs to suppress Plasmodium berghei blood-stage parasite proliferation. Resistance selection studies, confirmed by CRISPR-Cas9-based gene editing, identified the digestive vacuole membrane-spanning transporter PfMDR1 (P. falciparum multidrug resistance gene-1) as a determinant of parasite resistance to HHQs. Haemoglobin and haem fractionation assays suggest a mode of action that results in reduced haemozoin levels and might involve inhibition of host haemoglobin uptake into intra-erythrocytic parasites. Furthermore, parasites resistant to HHQs displayed increased susceptibility to several first-line antimalarial drugs, including lumefantrine, confirming that HHQs have a different mode of action to other antimalarials drugs for which PfMDR1 is known to confer resistance. This work evokes therapeutic strategies that combine opposing selective pressures on this parasite transporter as an approach to countering the emergence and transmission of multidrug-resistant P. falciparum malaria.
Collapse
Affiliation(s)
- Manu Vanaerschot
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Leonardo Lucantoni
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, 4111, Queensland, Australia
| | - Tao Li
- Sanaria Inc., Rockville, MD, 20852, USA
| | - Jill M Combrinck
- Division of Pharmacology, Department of Medicine, University of Cape Town, Cape Town, 7925, South Africa
| | - Andrea Ruecker
- Department of Life Sciences, Imperial College, London, SW7 2AZ, UK.,Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand.,Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7FZ, UK
| | - T R Santha Kumar
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Kelly Rubiano
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Pedro E Ferreira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal
| | - Giulia Siciliano
- Dipartimento di Malattie Infettive, Parassitarie ed Immunomediate, Istituto Superiore di Sanità, 00161, Rome, Italy
| | - Sonia Gulati
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Philipp P Henrich
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Caroline L Ng
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY, 10032, USA
| | - James M Murithi
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Victoria C Corey
- University of California, San Diego, School of Medicine, La Jolla, CA, 92093, USA
| | - Sandra Duffy
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, 4111, Queensland, Australia
| | - Ori J Lieberman
- Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY, 10032, USA
| | - M Isabel Veiga
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057, Braga, Portugal
| | - Robert E Sinden
- Department of Life Sciences, Imperial College, London, SW7 2AZ, UK
| | - Pietro Alano
- Dipartimento di Malattie Infettive, Parassitarie ed Immunomediate, Istituto Superiore di Sanità, 00161, Rome, Italy
| | - Michael J Delves
- Department of Life Sciences, Imperial College, London, SW7 2AZ, UK
| | | | - Elizabeth A Winzeler
- University of California, San Diego, School of Medicine, La Jolla, CA, 92093, USA
| | - Timothy J Egan
- Department of Chemistry, University of Cape Town, Cape Town, 7700, South Africa
| | | | - Vicky M Avery
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, 4111, Queensland, Australia
| | - 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
|
175
|
Antimalarial Resistance Unlikely To Explain U.K. Artemether-Lumefantrine Failures. Antimicrob Agents Chemother 2017; 61:61/7/e00721-17. [PMID: 28655744 DOI: 10.1128/aac.00721-17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
176
|
Nag S, Dalgaard MD, Kofoed PE, Ursing J, Crespo M, Andersen LO, Aarestrup FM, Lund O, Alifrangis M. High throughput resistance profiling of Plasmodium falciparum infections based on custom dual indexing and Illumina next generation sequencing-technology. Sci Rep 2017; 7:2398. [PMID: 28546554 PMCID: PMC5445084 DOI: 10.1038/s41598-017-02724-x] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 04/18/2017] [Indexed: 01/10/2023] Open
Abstract
Genetic polymorphisms in P. falciparum can be used to indicate the parasite's susceptibility to antimalarial drugs as well as its geographical origin. Both of these factors are key to monitoring development and spread of antimalarial drug resistance. In this study, we combine multiplex PCR, custom designed dual indexing and Miseq sequencing for high throughput SNP-profiling of 457 malaria infections from Guinea-Bissau, at the cost of 10 USD per sample. By amplifying and sequencing 15 genetic fragments, we cover 20 resistance-conferring SNPs occurring in pfcrt, pfmdr1, pfdhfr, pfdhps, as well as the entire length of pfK13, and the mitochondrial barcode for parasite origin. SNPs of interest were sequenced with an average depth of 2,043 reads, and bases were called for the various SNP-positions with a p-value below 0.05, for 89.8-100% of samples. The SNP data indicates that artemisinin resistance-conferring SNPs in pfK13 are absent from the studied area of Guinea-Bissau, while the pfmdr1 86 N allele is found at a high prevalence. The mitochondrial barcodes are unanimous and accommodate a West African origin of the parasites. With this method, very reliable high throughput surveillance of antimalarial drug resistance becomes more affordable than ever before.
Collapse
Affiliation(s)
- Sidsel Nag
- Centre for Medical Parasitology, Department of International Health, Immunology and Microbiology, University of Copenhagen, 1356, Copenhagen K, Denmark.
- Department of Infectious Diseases, Copenhagen University Hospital, 2200, Copenhagen N, Denmark.
| | - Marlene D Dalgaard
- Department of Systems Biology, Technical University of Denmark, Kemitorvet Building 208, 2800, Kgs. Lyngby, Denmark
| | - Poul-Erik Kofoed
- Department of Paediatrics, Kolding Hospital, University of Southern Denmark, 6000, Kolding, Denmark
- Bandim Health Project, Bissau, Guinea-Bissau
| | - Johan Ursing
- Bandim Health Project, Bissau, Guinea-Bissau
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Marina Crespo
- Centre for Medical Parasitology, Department of International Health, Immunology and Microbiology, University of Copenhagen, 1356, Copenhagen K, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, 2200, Copenhagen N, Denmark
| | - Lee O'Brien Andersen
- Department of Microbiology and Infection Control, Statens Serum Institut, 2300, Copenhagen S, Denmark
| | | | - Ole Lund
- Department of Systems Biology, Technical University of Denmark, Kemitorvet Building 208, 2800, Kgs. Lyngby, Denmark
| | - Michael Alifrangis
- Centre for Medical Parasitology, Department of International Health, Immunology and Microbiology, University of Copenhagen, 1356, Copenhagen K, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, 2200, Copenhagen N, Denmark
| |
Collapse
|
177
|
Nguetse CN, Adegnika AA, Agbenyega T, Ogutu BR, Krishna S, Kremsner PG, Velavan TP. Molecular markers of anti-malarial drug resistance in Central, West and East African children with severe malaria. Malar J 2017; 16:217. [PMID: 28535801 PMCID: PMC5442681 DOI: 10.1186/s12936-017-1868-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 05/17/2017] [Indexed: 11/10/2022] Open
Abstract
Background The Plasmodium falciparum multidrug resistance 1 (PfMDR1), P. falciparum Ca2+-ATPase (PfATP6) and Kelch-13 propeller domain (PfK13) loci are molecular markers of parasite susceptibility to anti-malarial drugs. Their frequency distributions were determined in the isolates collected from children with severe malaria originating from three African countries. Methods Samples from 287 children with severe malaria [(Gabon: n = 114); (Ghana: n = 89); (Kenya: n = 84)] were genotyped for pfmdr1, pfatp6 and pfk13 loci by DNA sequencing and assessing pfmdr1 copy number variation (CNV) by real-time PCR. Results Pfmdr1-N86Y mutation was detected in 48, 10 and 10% in Lambaréné, Kumasi and Kisumu, respectively. At codon 184, the prevalence of the mutation was 73% in Lambaréné, 63% in Kumasi and 49% Kisumu. The S1034C and N1042D variants were absent at all three sites, while the frequency of the D1246Y mutation was 1, 3 and 13% in Lambaréné, Kumasi and Kisumu, respectively. Isolates with two pfmdr1 gene copy number predominantly harboured the N86Y wild-type allele and were mostly found in Kumasi (10%) (P < 0.0001). Among the main pfmdr1 haplotypes (NFD, NYD and YFD), NYD was associated with highest parasitaemia (P = 0.04). At the pfatp6 locus, H243Y and A623E mutations were observed at very low frequency at all three sites. The prevalence of the pfatp6 E431K variant was 6, 18 and 17% in Lambaréné, Kumasi and Kisumu, respectively. The L263E and S769N mutations were absent in all isolates. The pfk13 variants associated with artemisinin resistance in Southeast Asia were not observed. Eleven novel substitutions in the pfk13 locus occurring at low frequency were observed. Conclusions Artemisinins are still highly efficacious in large malaria-endemic regions though declining efficacy has occurred in Southeast Asia. The return of chloroquine-sensitive strains following the removal of drug pressure is observed. However, selection of wild-type alleles in the multidrug-resistance gene and the increased gene copy number is associated with reduced lumefantrine sensitivity. This study indicates a need to constantly monitor drug resistance to artemisinin in field isolates from malaria-endemic countries.
Collapse
Affiliation(s)
- Christian N Nguetse
- Institute of Tropical Medicine, University Tübingen, Wilhelmstrasse 27, 72074, Tübingen, Germany
| | - Ayola Akim Adegnika
- Institute of Tropical Medicine, University Tübingen, Wilhelmstrasse 27, 72074, Tübingen, Germany.,Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| | - Tsiri Agbenyega
- Department of Physiology, University of Science and Technology, School of Medical Sciences, Kumasi, Ghana.,Departments of Child Health and Medicine, Komfo Anokye Teaching Hospital, Kumasi, Ghana
| | - Bernhards R Ogutu
- Centre for Clinical Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Sanjeev Krishna
- Institute of Tropical Medicine, University Tübingen, Wilhelmstrasse 27, 72074, Tübingen, Germany.,Institute for Infection and Immunity, St George's University of London, London, UK
| | - Peter G Kremsner
- Institute of Tropical Medicine, University Tübingen, Wilhelmstrasse 27, 72074, Tübingen, Germany.,Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| | - Thirumalaisamy P Velavan
- Institute of Tropical Medicine, University Tübingen, Wilhelmstrasse 27, 72074, Tübingen, Germany. .,Fondation Congolaise pour la Recherche Médicale, Brazzaville, Republic of Congo. .,Vietnamese-German Center for Medical Research, Hanoi, Vietnam. .,Faculty of Medicine, Duy Tan University, Da Nang, Vietnam.
| |
Collapse
|
178
|
Crawford ED, Quan J, Horst JA, Ebert D, Wu W, DeRisi JL. Plasmid-free CRISPR/Cas9 genome editing in Plasmodium falciparum confirms mutations conferring resistance to the dihydroisoquinolone clinical candidate SJ733. PLoS One 2017; 12:e0178163. [PMID: 28542423 PMCID: PMC5439709 DOI: 10.1371/journal.pone.0178163] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 05/08/2017] [Indexed: 12/31/2022] Open
Abstract
Genetic manipulation of the deadly malaria parasite Plasmodium falciparum remains challenging, but the rise of CRISPR/Cas9-based genome editing tools is increasing the feasibility of altering this parasite’s genome in order to study its biology. Of particular interest is the investigation of drug targets and drug resistance mechanisms, which have major implications for fighting malaria. We present a new method for introducing drug resistance mutations in P. falciparum without the use of plasmids or the need for cloning homologous recombination templates. We demonstrate this method by introducing edits into the sodium efflux channel PfATP4 by transfection of a purified CRISPR/Cas9-guide RNA ribonucleoprotein complex and a 200-nucleotide single-stranded oligodeoxynucleotide (ssODN) repair template. Analysis of whole genome sequencing data with the variant-finding program MinorityReport confirmed that only the intended edits were made, and growth inhibition assays confirmed that these mutations confer resistance to the antimalarial SJ733. The method described here is ideally suited for the introduction of mutations that confer a fitness advantage under selection conditions, and the novel finding that an ssODN can function as a repair template in P. falciparum could greatly simplify future editing attempts regardless of the nuclease used or the delivery method.
Collapse
Affiliation(s)
- Emily D. Crawford
- Chan Zuckerberg Biohub, San Francisco, California, United States of America
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, California, United States of America
- Howard Hughes Medical Institute, Chevy Chase, Maryland, United States of America
| | - Jenai Quan
- Chan Zuckerberg Biohub, San Francisco, California, United States of America
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, California, United States of America
- Howard Hughes Medical Institute, Chevy Chase, Maryland, United States of America
| | - Jeremy A. Horst
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, California, United States of America
| | - Daniel Ebert
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, California, United States of America
- Howard Hughes Medical Institute, Chevy Chase, Maryland, United States of America
| | - Wesley Wu
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, California, United States of America
| | - Joseph L. DeRisi
- Chan Zuckerberg Biohub, San Francisco, California, United States of America
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, California, United States of America
- Howard Hughes Medical Institute, Chevy Chase, Maryland, United States of America
- * E-mail:
| |
Collapse
|
179
|
A Variant PfCRT Isoform Can Contribute to Plasmodium falciparum Resistance to the First-Line Partner Drug Piperaquine. mBio 2017; 8:mBio.00303-17. [PMID: 28487425 PMCID: PMC5424201 DOI: 10.1128/mbio.00303-17] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Current efforts to reduce the global burden of malaria are threatened by the rapid spread throughout Asia of Plasmodium falciparum resistance to artemisinin-based combination therapies, which includes increasing rates of clinical failure with dihydroartemisinin plus piperaquine (PPQ) in Cambodia. Using zinc finger nuclease-based gene editing, we report that addition of the C101F mutation to the chloroquine (CQ) resistance-conferring PfCRT Dd2 isoform common to Asia can confer PPQ resistance to cultured parasites. Resistance was demonstrated as significantly higher PPQ concentrations causing 90% inhibition of parasite growth (IC90) or 50% parasite killing (50% lethal dose [LD50]). This mutation also reversed Dd2-mediated CQ resistance, sensitized parasites to amodiaquine, quinine, and artemisinin, and conferred amantadine and blasticidin resistance. Using heme fractionation assays, we demonstrate that PPQ causes a buildup of reactive free heme and inhibits the formation of chemically inert hemozoin crystals. Our data evoke inhibition of heme detoxification in the parasite’s acidic digestive vacuole as the primary mode of both the bis-aminoquinoline PPQ and the related 4-aminoquinoline CQ. Both drugs also inhibit hemoglobin proteolysis at elevated concentrations, suggesting an additional mode of action. Isogenic lines differing in their pfmdr1 copy number showed equivalent PPQ susceptibilities. We propose that mutations in PfCRT could contribute to a multifactorial basis of PPQ resistance in field isolates. The global agenda to eliminate malaria depends on the continued success of artemisinin-based combination therapies (ACTs), which target the asexual blood stages of the intracellular parasite Plasmodium. Partial resistance to artemisinin, however, is now established in Southeast Asia, exposing the partner drugs to increased selective pressure. Plasmodium falciparum resistance to the first-line partner piperaquine (PPQ) is now spreading rapidly in Cambodia, resulting in clinical treatment failures. Here, we report that a variant form of the Plasmodium falciparum chloroquine resistance transporter, harboring a C101F mutation edited into the chloroquine (CQ)-resistant Dd2 isoform prevalent in Asia, can confer PPQ resistance in cultured parasites. This was accompanied by a loss of CQ resistance. Biochemical assays showed that PPQ, like CQ, inhibits the detoxification of reactive heme that is formed by parasite-mediated catabolism of host hemoglobin. We propose that novel PfCRT variants emerging in the field could contribute to a multigenic basis of PPQ resistance.
Collapse
|
180
|
Pholwat S, Liu J, Stroup S, Jacob ST, Banura P, Moore CC, Huang F, Laufer MK, Houpt E, Guler JL. The Malaria TaqMan Array Card Includes 87 Assays for Plasmodium falciparum Drug Resistance, Identification of Species, and Genotyping in a Single Reaction. Antimicrob Agents Chemother 2017; 61:e00110-17. [PMID: 28264857 PMCID: PMC5404514 DOI: 10.1128/aac.00110-17] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 03/02/2017] [Indexed: 11/20/2022] Open
Abstract
Antimalarial drug resistance exacerbates the global disease burden and complicates eradication efforts. To facilitate the surveillance of resistance markers in countries of malaria endemicity, we developed a suite of TaqMan assays for known resistance markers and compartmentalized them into a single array card (TaqMan array card, TAC). We included 87 assays for species identification, for the detection of Plasmodium falciparum mutations associated with chloroquine, atovaquone, pyrimethamine, sulfadoxine, and artemisinin resistance, and for neutral single nucleotide polymorphism (SNP) genotyping. Assay performance was first optimized using DNA from common laboratory parasite lines and plasmid controls. The limit of detection was 0.1 to 10 pg of DNA and yielded 100% accuracy compared to sequencing. The tool was then evaluated on 87 clinical blood samples from around the world, and the malaria TAC once again achieved 100% accuracy compared to sequencing and in addition detected the presence of mixed infections in clinical samples. With its streamlined protocol and high accuracy, this malaria TAC should be a useful tool for large-scale antimalarial resistance surveillance.
Collapse
Affiliation(s)
- Suporn Pholwat
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Jie Liu
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Suzanne Stroup
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Shevin T Jacob
- Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington, USA
| | - Patrick Banura
- Department of Community Health, Masaka Regional Referral Hospital, Masaka, Uganda
| | - Christopher C Moore
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Fang Huang
- Division of Malaria Research, Institute for Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, China
| | - Miriam K Laufer
- Division of Malaria Research, Institute for Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Eric Houpt
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Jennifer L Guler
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, Virginia, USA
- Department of Biology, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
181
|
Koukouikila-Koussounda F, Jeyaraj S, Nguetse CN, Nkonganyi CN, Kokou KC, Etoka-Beka MK, Ntoumi F, Velavan TP. Molecular surveillance of Plasmodium falciparum drug resistance in the Republic of Congo: four and nine years after the introduction of artemisinin-based combination therapy. Malar J 2017; 16:155. [PMID: 28420403 PMCID: PMC5395861 DOI: 10.1186/s12936-017-1816-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 04/09/2017] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Resistance to anti-malarial drugs hinders efforts on malaria elimination and eradication. Following the global spread of chloroquine-resistant parasites, the Republic of Congo adopted artemisinin-based combination therapy (ACT) in 2006 as a first-line treatment for uncomplicated malaria. To assess the impacts after implementation of ACT, a molecular surveillance for anti-malarial drug resistance was conducted in Congo 4 and 9 years after the introduction of ACT. METHODS Blood samples of 431 febrile children aged 1-10 years were utilized from two previous studies conducted in 2010 (N = 311) and 2015 (N = 120). All samples were screened for malaria parasites using nested PCR. Direct sequencing was used to determine the frequency distribution of genetic variants in the anti-malarial drug-resistant Plasmodium falciparum genes (Pfcrt, Pfmdr1, Pfatp6, Pfk13) in malaria-positive isolates. RESULTS One-hundred and nineteen (N = 70 from 2010 and N = 49 from 2015) samples were positive for P. falciparum. A relative decrease in the proportion of chloroquine-resistant haplotype (CVIET) from 100% in 2005, 1 year before the introduction and implementation of ACT in 2006, to 98% in 2010 to 71% in 2015 was observed. Regarding the multidrug transporter gene, a considerable reduction in the frequency of the mutations N86Y (from 73 to 27%) and D1246Y (from 22 to 0%) was observed. However, the prevalence of the Y184F mutation remained stable (49% in 2010 compared to 54% in 2015). Isolates carrying the Pfatp6 H243Y was 25% in 2010 and this frequency was reduced to null in 2015. None of the parasites harboured the Pfk13 mutations associated with prolonged artemisinin clearance in Southeast Asia. Nevertheless, 13 new Pfk13 variants are reported among the investigated isolates. CONCLUSION The implementation of ACT has led to the decline in prevalence of chloroquine-resistant parasites in the Republic of Congo. However, the constant prevalence of the PfMDR1 Y184F mutation, associated with lumefantrine susceptibility, indicate a selective drug pressure still exists. Taken together, this study could serve as the basis for epidemiological studies monitoring the distribution of molecular markers of artemisinin resistance in the Republic of Congo.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Francine Ntoumi
- Fondation Congolaise pour la Recherche Médicale (FCRM), Brazzaville, Republic of Congo.,Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
| | - Thirumalaisamy P Velavan
- Fondation Congolaise pour la Recherche Médicale (FCRM), Brazzaville, Republic of Congo. .,Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany. .,Duy Tan University, Da Nang, Vietnam. .,Vietnamese-German Centre for Medical Research, Hanoi, Vietnam.
| |
Collapse
|
182
|
Costa GL, Amaral LC, Fontes CJF, Carvalho LH, de Brito CFA, de Sousa TN. Assessment of copy number variation in genes related to drug resistance in Plasmodium vivax and Plasmodium falciparum isolates from the Brazilian Amazon and a systematic review of the literature. Malar J 2017; 16:152. [PMID: 28420389 PMCID: PMC5395969 DOI: 10.1186/s12936-017-1806-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 04/07/2017] [Indexed: 12/29/2022] Open
Abstract
Background Parasite resistance to anti-malarials represents a great obstacle for malaria elimination. The majority of studies have investigated the association between single-nucleotide polymorphisms (SNPs) and drug resistance; however, it is becoming clear that the copy number variation (CNV) is also associated with this parasite phenotype. To provide a baseline for molecular surveillance of anti-malarial drug resistance in the Brazilian Amazon, the present study characterized the genetic profile of both markers in the most common genes associated with drug resistance in Plasmodium falciparum and Plasmodium vivax isolates. Additionally, these data were compared to data published elsewhere applying a systematic review of the literature published over a 20-year time period. Methods The genomic DNA of 67 patients infected by P. falciparum and P. vivax from three Brazilian States was obtained between 2002 and 2012. CNV in P. falciparum multidrug resistance gene-1 (pfmdr1), GTP cyclohydrolase 1 (pfgch1) and P. vivax multidrug resistance gene-1 (pvmdr1) were assessed by real-time PCR assays. SNPs in the pfmdr1 and pfcrt genes were assessed by PCR–RFLP. A literature search for studies that analysed CNP in the same genes of P. falciparum and P. vivax was conducted between May 2014 and March 2017 across four databases. Results All analysed samples of P. falciparum carried only one copy of pfmdr1 or pfgch1. Although the pfcrt K76T polymorphism, a determinant of CQ resistance, was present in all samples genotyped, the pfmdr1 N86Y was absent. For P. vivax isolates, an amplification rate of 20% was found for the pvmdr1 gene. The results of the study are in agreement with the low amplification rates for pfmdr1 gene evidenced in the Americas and Africa, while higher rates have been described in Southeast Asia. For P. vivax, very low rates of amplification for pvmdr1 have been described worldwide, with exceptions in French Guiana, Cambodia, Thailand and Brazil. Conclusions The present study was the first to evaluate gch1 CNV in P. falciparum isolates from Brazil, showing an absence of amplification of this gene more than 20 years after the withdrawal of the Brazilian antifolates therapeutic scheme. Furthermore, the rate of pvmdr1 amplification was significantly higher than that previously reported for isolates circulating in Northern Brazil. Electronic supplementary material The online version of this article (doi:10.1186/s12936-017-1806-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Gabriel Luíz Costa
- Molecular Biology and Malaria Immunology Research Group, Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz (FIOCRUZ), Belo Horizonte, Minas Gerais, Brazil
| | - Lara Cotta Amaral
- Molecular Biology and Malaria Immunology Research Group, Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz (FIOCRUZ), Belo Horizonte, Minas Gerais, Brazil
| | | | - Luzia Helena Carvalho
- Molecular Biology and Malaria Immunology Research Group, Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz (FIOCRUZ), Belo Horizonte, Minas Gerais, Brazil
| | - Cristiana Ferreira Alves de Brito
- Molecular Biology and Malaria Immunology Research Group, Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz (FIOCRUZ), Belo Horizonte, Minas Gerais, Brazil
| | - Taís Nóbrega de Sousa
- Molecular Biology and Malaria Immunology Research Group, Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz (FIOCRUZ), Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
183
|
Gil JP, Krishna S. pfmdr1 (Plasmodium falciparum multidrug drug resistance gene 1): a pivotal factor in malaria resistance to artemisinin combination therapies. Expert Rev Anti Infect Ther 2017; 15:527-543. [DOI: 10.1080/14787210.2017.1313703] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- J. Pedro Gil
- Physiology and Pharmacology Department, Karolinska Institutet, Stockholm, Sweden
| | - S. Krishna
- St George’s University Hospital, Institute for Infection and Immunity, London, United Kingdom
| |
Collapse
|
184
|
Mungthin M, Watanatanasup E, Sitthichot N, Suwandittakul N, Khositnithikul R, Ward SA. Influence of the pfmdr1 Gene on In Vitro Sensitivities of Piperaquine in Thai Isolates of Plasmodium falciparum. Am J Trop Med Hyg 2017; 96:624-629. [PMID: 28044042 DOI: 10.4269/ajtmh.16-0668] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Piperaquine combined with dihydroartemisinin is one of the artemisinin derivative combination therapies, which can replace artesunate-mefloquine in treating uncomplicated falciparum malaria in Thailand. The aim of this study was to determine the in vitro sensitivity of Thai Plasmodium falciparum isolates against piperaquine and the influence of the pfmdr1 gene on in vitro response. One hundred and thirty-seven standard laboratory and adapted Thai isolates of P. falciparum were assessed for in vitro piperaquine sensitivity. Polymorphisms of the pfmdr1 gene were determined by polymerase chain reaction methods. The mean and standard deviation of the piperaquine IC50 in Thai isolates of P. falciparum were 16.7 ± 6.3 nM. The parasites exhibiting chloroquine IC50 of ≥ 100 nM were significantly less sensitive to piperaquine compared with the parasite with chloroquine IC50 of < 100 nM. No significant association between the pfmdr1 copy number and piperaquine IC50 values was found. In contrast, the parasites containing the pfmdr1 86Y allele exhibited significantly reduced piperaquine sensitivity. Before nationwide implementation of dihydroartemisinin-piperaquine as the first-line treatment in Thailand, in vitro and in vivo evaluations of this combination should be performed especially in areas where parasites containing the pfmdr1 86Y allele are predominant such as the Thai-Malaysian border.
Collapse
Affiliation(s)
- Mathirut Mungthin
- Department of Parasitology, Phramongkutklao College of Medicine, Bangkok, Thailand
| | | | - Naruemon Sitthichot
- Department of Parasitology, Phramongkutklao College of Medicine, Bangkok, Thailand
| | | | | | - Stephen A Ward
- Division of Molecular and Biochemical Parasitology, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| |
Collapse
|
185
|
The "pushmi-pullyu" of resistance to chloroquine in malaria. Essays Biochem 2017; 61:167-175. [PMID: 28258239 DOI: 10.1042/ebc20160060] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 12/27/2016] [Accepted: 01/03/2017] [Indexed: 11/17/2022]
Abstract
Malarial infection continues to impart devastating health problems in the developing world. Treatment of malaria has involved chemotherapy since 168 BC, with the most prevalent and successful forms using plant alkaloids. Perhaps the greatest treatment success against malaria was by chloroquine, a synthetic derivative of the quinines found in the Cinchona tree bark. Chloroquine is able to kill parasites by interfering with haem metabolism in the parasite's digestive vacuole. The widespread use of chloroquine predictably resulted in the development of drug-resistant malaria and the most highly implicated resistance mediators are the transporter proteins P-glycoprotein (P-gp) homologue 1 (P-gh1) and Plasmodium falciparum chloroquine-resistance transporter (PfCRT), which reside on the parasite's digestive vacuole. The presence of PfCRT and P-gh1 on the vacuole membrane is analogous to the two-headed fictional creature known as the "Pushmi-Pullyu". P-gh1 (Pushmi) increases influx of chloroquine into the vacuole, while PfCRT (Pullmi) causes efflux of chloroquine from the vacuole. This review describes how drug-resistant malarial parasites co-ordinate chloroquine distribution through adaptive mutations to promote their survival in the presence of this cytotoxic drug.
Collapse
|
186
|
Burle-Caldas GA, Grazielle-Silva V, Soares-Simões M, Schumann Burkard G, Roditi I, DaRocha WD, Teixeira SM. Editing the Trypanosoma cruzi genome with zinc finger nucleases. Mol Biochem Parasitol 2017; 212:28-32. [DOI: 10.1016/j.molbiopara.2017.01.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 01/13/2017] [Accepted: 01/13/2017] [Indexed: 01/23/2023]
|
187
|
Evolution of Fitness Cost-Neutral Mutant PfCRT Conferring P. falciparum 4-Aminoquinoline Drug Resistance Is Accompanied by Altered Parasite Metabolism and Digestive Vacuole Physiology. PLoS Pathog 2016; 12:e1005976. [PMID: 27832198 PMCID: PMC5104409 DOI: 10.1371/journal.ppat.1005976] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 10/03/2016] [Indexed: 11/19/2022] Open
Abstract
Southeast Asia is an epicenter of multidrug-resistant Plasmodium falciparum strains. Selective pressures on the subcontinent have recurrently produced several allelic variants of parasite drug resistance genes, including the P. falciparum chloroquine resistance transporter (pfcrt). Despite significant reductions in the deployment of the 4-aminoquinoline drug chloroquine (CQ), which selected for the mutant pfcrt alleles that halted CQ efficacy decades ago, the parasite pfcrt locus is continuously evolving. This is highlighted by the presence of a highly mutated allele, Cam734 pfcrt, which has acquired the singular ability to confer parasite CQ resistance without an associated fitness cost. Here, we used pfcrt-specific zinc-finger nucleases to genetically dissect this allele in the pathogenic setting of asexual blood-stage infection. Comparative analysis of drug resistance and growth profiles of recombinant parasites that express Cam734 or variants thereof, Dd2 (the most common Southeast Asian variant), or wild-type pfcrt, revealed previously unknown roles for PfCRT mutations in modulating parasite susceptibility to multiple antimalarial agents. These results were generated in the GC03 strain, used in multiple earlier pfcrt studies, and might differ in natural isolates harboring this allele. Results presented herein show that Cam734-mediated CQ resistance is dependent on the rare A144F mutation that has not been observed beyond Southeast Asia, and reveal distinct impacts of this and other Cam734-specific mutations on CQ resistance and parasite growth rates. Biochemical assays revealed a broad impact of mutant PfCRT isoforms on parasite metabolism, including nucleoside triphosphate levels, hemoglobin catabolism and disposition of heme, as well as digestive vacuole volume and pH. Results from our study provide new insights into the complex molecular basis and physiological impact of PfCRT-mediated antimalarial drug resistance, and inform ongoing efforts to characterize novel pfcrt alleles that can undermine the efficacy of first-line antimalarial drug regimens. Point mutations in the Plasmodium falciparum chloroquine resistance transporter (PfCRT) earlier thwarted the clinical efficacy of chloroquine, the former gold standard, and constitute a major determinant of parasite susceptibility to antimalarial drugs. Recently, we reported that the highly mutated Cambodian PfCRT isoform Cam734 is fitness-neutral in terms of parasite growth, unlike other less fit isoforms such as Dd2 that are outcompeted by wild-type parasites in the absence of CQ pressure. Using pfcrt-specific zinc-finger nucleases to genetically dissect the Cam734 allele, we report that its unique constituent mutations directly contribute to CQ resistance and collectively offset fitness costs associated with intermediate mutational steps. We also report that these mutations can contribute to resistance or increased sensitivity to multiple first-line partner drugs. Using isogenic parasite lines, we provide evidence of changes in parasite metabolism associated with the Cam734 allele compared to Dd2. We also observe a close correlation between CQ inhibition of hemozoin formation and parasite growth, and provide evidence that Cam734 PfCRT can modulate drug potency depending on its membrane electrochemical gradient. Our data highlight the capacity of PfCRT to evolve new states of antimalarial drug resistance and to offset associated fitness costs through its impact on parasite physiology and hemoglobin catabolism.
Collapse
|
188
|
Srimuang K, Miotto O, Lim P, Fairhurst RM, Kwiatkowski DP, Woodrow CJ, Imwong M. Analysis of anti-malarial resistance markers in pfmdr1 and pfcrt across Southeast Asia in the Tracking Resistance to Artemisinin Collaboration. Malar J 2016; 15:541. [PMID: 27825353 PMCID: PMC5101715 DOI: 10.1186/s12936-016-1598-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Accepted: 10/31/2016] [Indexed: 01/07/2023] Open
Abstract
Background Declining anti-malarial efficacy of artemisinin-based combination therapy, and reduced Plasmodium falciparum susceptibility to individual anti-malarials are being documented across an expanding area of Southeast Asia (SEA). Genotypic markers complement phenotypic studies in assessing the efficacy of individual anti-malarials. Methods The markers pfmdr1 and pfcrt were genotyped in parasite samples obtained in 2011–2014 at 14 TRAC (Tracking Resistance to Artemisinin Collaboration) sites in mainland Southeast Asia using a combination of PCR and next-generation sequencing methods. Results Pfmdr1 amplification, a marker of mefloquine and lumefantrine resistance, was highly prevalent at Mae Sot on the Thailand–Myanmar border (59.8% of isolates) and common (more than 10%) at sites in central Myanmar, eastern Thailand and western Cambodia; however, its prevalence was lower than previously documented in Pailin, western Cambodia. The pfmdr1 Y184F mutation was common, particularly in and around Cambodia, and the F1226Y mutation was found in about half of samples in Mae Sot. The functional significance of these two mutations remains unclear. Other previously documented pfmdr1 mutations were absent or very rare in the region. The pfcrt mutation K76T associated with chloroquine resistance was found in 98.2% of isolates. The CVIET haplotype made up 95% or more of isolates in western SEA while the CVIDT haplotype was common (30–40% of isolates) in north and northeastern Cambodia, southern Laos, and southern Vietnam. Conclusions These findings generate cause for concern regarding the mid-term efficacy of artemether–lumefantrine in Myanmar, while the absence of resistance-conferring pfmdr1 mutations and SVMNT pfcrt haplotypes suggests that amodiaquine could be an efficacious component of anti-malarial regimens in SEA. Electronic supplementary material The online version of this article (doi:10.1186/s12936-016-1598-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Krongkan Srimuang
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Olivo Miotto
- Mahidol Oxford Tropical Medicine Research Unit, Bangkok, Thailand.,Wellcome Trust Sanger Institute, Hinxton, UK.,Medical Research Council (MRC) Centre for Genomics and Global Health, University of Oxford, Oxford, UK
| | - Pharath Lim
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20852, USA
| | - Rick M Fairhurst
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, 20852, USA
| | - Dominic P Kwiatkowski
- Wellcome Trust Sanger Institute, Hinxton, UK.,Medical Research Council (MRC) Centre for Genomics and Global Health, University of Oxford, Oxford, UK
| | - Charles J Woodrow
- Mahidol Oxford Tropical Medicine Research Unit, Bangkok, Thailand.,Centre for Tropical Medicine & Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Mallika Imwong
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand. .,Mahidol Oxford Tropical Medicine Research Unit, Bangkok, Thailand.
| | | |
Collapse
|
189
|
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: 258] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [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
|
190
|
Volkman SK, Herman J, Lukens AK, Hartl DL. Genome-Wide Association Studies of Drug-Resistance Determinants. Trends Parasitol 2016; 33:214-230. [PMID: 28179098 DOI: 10.1016/j.pt.2016.10.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 09/26/2016] [Accepted: 10/06/2016] [Indexed: 02/07/2023]
Abstract
Population genetic strategies that leverage association, selection, and linkage have identified drug-resistant loci. However, challenges and limitations persist in identifying drug-resistance loci in malaria. In this review we discuss the genetic basis of drug resistance and the use of genome-wide association studies, complemented by selection and linkage studies, to identify and understand mechanisms of drug resistance and response. We also discuss the implications of nongenetic mechanisms of drug resistance recently reported in the literature, and present models of the interplay between nongenetic and genetic processes that contribute to the emergence of drug resistance. Throughout, we examine artemisinin resistance as an example to emphasize challenges in identifying phenotypes suitable for population genetic studies as well as complications due to multiple-factor drug resistance.
Collapse
Affiliation(s)
- Sarah K Volkman
- Harvard T.H. Chan School of Public Health, Department of Immunology and Infectious Disease, Boston, MA, USA; The Broad Institute of MIT and Harvard, Infectious Disease Initiative, Cambridge, MA, USA; Simmons College, School of Nursing and Health Science, Boston, MA, USA.
| | - Jonathan Herman
- Harvard T.H. Chan School of Public Health, Department of Immunology and Infectious Disease, Boston, MA, USA; Weill Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Amanda K Lukens
- Harvard T.H. Chan School of Public Health, Department of Immunology and Infectious Disease, Boston, MA, USA; The Broad Institute of MIT and Harvard, Infectious Disease Initiative, Cambridge, MA, USA
| | - Daniel L Hartl
- The Broad Institute of MIT and Harvard, Infectious Disease Initiative, Cambridge, MA, USA; Harvard University, Organismic and Evolutionary Biology, Cambridge, MA, USA
| |
Collapse
|
191
|
Parallel inhibition of amino acid efflux and growth of erythrocytic Plasmodium falciparum by mefloquine and non-piperidine analogs: Implication for the mechanism of antimalarial action. Bioorg Med Chem Lett 2016; 26:4846-4850. [DOI: 10.1016/j.bmcl.2016.08.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 08/01/2016] [Accepted: 08/02/2016] [Indexed: 11/18/2022]
|
192
|
Chatterjee M, Ganguly S, Saha P, Guha SK, Basu N, Bera DK, Maji AK. Polymorphisms in Pfcrt and Pfmdr-1 genes after five years withdrawal of chloroquine for the treatment of Plasmodium falciparum malaria in West Bengal, India. INFECTION GENETICS AND EVOLUTION 2016; 44:281-285. [PMID: 27448953 DOI: 10.1016/j.meegid.2016.07.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 07/11/2016] [Accepted: 07/19/2016] [Indexed: 12/29/2022]
Abstract
BACKGROUND The emergence of resistant power against different antimalarial agents particularly by Plasmodium falciparum is a challenge to combat malaria. Regular monitoring is essential not only to determine the efficacy and development of resistance by the parasite but also to detect early sign of regaining sensitivity to any anti-malarial agent that has been withdrawn for a long period. Studies on molecular markers associated with antimalarial drug resistance of prevailing Plasmodium population play an important role in this aspect. The present protocol was designed to study the polymorphisms in pfcrt and pfmdr-1 gene to determine any sign of regaining sensitivity to chloroquine among P. falciparum after five years of artemisinin combination therapy (ACT) implementation. METHODS Clinical isolates were collected from P. falciparum positive patients attending the malaria clinic of Calcutta School of Tropical Medicine during December 2014 to December 2015. Genomic parasitic DNA was extracted and subjected to sequencing of pfcrt and pfmdr-1 gene directly from purified PCR products. RESULTS A total of 89 isolates were sequenced for pfcrt and 73 isolates for pfmdr-1 genes. In pfcrt gene mutant K76T was detected in all isolates and all were SVMNT haplotype. Out of three important polymorphisms in pfmdr-1 gene mutant Y184F was detected among all isolates. One synonymous G182G and one non-synonymous S232F/Y, mutation were detected in 99% isolates. CONCLUSION All isolates carrying mutant K76T in pfcrt gene, considered as hall mark for CQ resistance, indicate that there is no sign of regaining CQ sensitivity among the prevailing P. falciparum population of the study area after five years of ACT implementation.
Collapse
Affiliation(s)
- Moytrey Chatterjee
- Protozoology Unit, Department of Microbiology, Calcutta School of Tropical Medicine, 108, C. R. Avenue, Kolkata 700 073, India.
| | - Swagata Ganguly
- Department of Microbiology, NRS Medical College, 138 AJC Bose Road, Kolkata 700 014, India.
| | - Pabitra Saha
- Department of Zoology, APC Roy Government College, Himachal Bihar, Matigara, Siliguri 734 010, West Bengal, India.
| | - Subhasish Kamal Guha
- Department of Tropical Medicine, Calcutta School of Tropical Medicine, 108, C. R. Avenue, Kolkata 700 073, India.
| | - Nandita Basu
- Calcutta School of Tropical Medicine, 108, C. R. Avenue, Kolkata 700 073, India.
| | - Dilip K Bera
- Protozoology Unit, Department of Microbiology, Calcutta School of Tropical Medicine, 108, C. R. Avenue, Kolkata 700 073, India.
| | - Ardhendu Kumar Maji
- Protozoology Unit, Department of Microbiology, Calcutta School of Tropical Medicine, 108, C. R. Avenue, Kolkata 700 073, India.
| |
Collapse
|
193
|
Tilley L, Straimer J, Gnädig NF, Ralph SA, Fidock DA. Artemisinin Action and Resistance in Plasmodium falciparum. Trends Parasitol 2016; 32:682-696. [PMID: 27289273 DOI: 10.1016/j.pt.2016.05.010] [Citation(s) in RCA: 221] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 05/10/2016] [Accepted: 05/13/2016] [Indexed: 12/16/2022]
Abstract
The worldwide use of artemisinin-based combination therapies (ACTs) has contributed in recent years to a substantial reduction in deaths resulting from Plasmodium falciparum malaria. Resistance to artemisinins, however, has emerged in Southeast Asia. Clinically, resistance is defined as a slower rate of parasite clearance in patients treated with an artemisinin derivative or an ACT. These slow clearance rates associate with enhanced survival rates of ring-stage parasites briefly exposed in vitro to dihydroartemisinin. We describe recent progress made in defining the molecular basis of artemisinin resistance, which has identified a primary role for the P. falciparum K13 protein. Using K13 mutations as molecular markers, epidemiological studies are now tracking the emergence and spread of artemisinin resistance. Mechanistic studies suggest potential ways to overcome resistance.
Collapse
Affiliation(s)
- Leann Tilley
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Melbourne, Australia.
| | - Judith Straimer
- Department of Microbiology and Immunology, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Nina F Gnädig
- Department of Microbiology and Immunology, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Stuart A Ralph
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Melbourne, Australia
| | - David A Fidock
- Department of Microbiology and Immunology, Columbia University College of Physicians and Surgeons, New York, NY, USA; Division of Infectious Diseases, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, USA.
| |
Collapse
|
194
|
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: 6.3] [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
|