1
|
Su X, Stadler RV, Xu F, Wu J. Malaria Genomics, Vaccine Development, and Microbiome. Pathogens 2023; 12:1061. [PMID: 37624021 PMCID: PMC10459703 DOI: 10.3390/pathogens12081061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/14/2023] [Accepted: 08/15/2023] [Indexed: 08/26/2023] Open
Abstract
Recent advances in malaria genetics and genomics have transformed many aspects of malaria research in areas of molecular evolution, epidemiology, transmission, host-parasite interaction, drug resistance, pathogenicity, and vaccine development. Here, in addition to introducing some background information on malaria parasite biology, parasite genetics/genomics, and genotyping methods, we discuss some applications of genetic and genomic approaches in vaccine development and in studying interactions with microbiota. Genetic and genomic data can be used to search for novel vaccine targets, design an effective vaccine strategy, identify protective antigens in a whole-organism vaccine, and evaluate the efficacy of a vaccine. Microbiota has been shown to influence disease outcomes and vaccine efficacy; studying the effects of microbiota in pathogenicity and immunity may provide information for disease control. Malaria genetics and genomics will continue to contribute greatly to many fields of malaria research.
Collapse
Affiliation(s)
- Xinzhuan Su
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA; (R.V.S.); (F.X.); (J.W.)
| | | | | | | |
Collapse
|
2
|
Biswas A, Raran-Kurussi S, Narayan A, Kar A, Chandra Mashurabad P, Bhattacharyya MK, Mandal K. Efficient refolding and functional characterization of PfAMA1(DI+DII) expressed in E. coli. Biochem Biophys Rep 2021; 26:100950. [PMID: 33665380 PMCID: PMC7907217 DOI: 10.1016/j.bbrep.2021.100950] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 01/18/2021] [Accepted: 02/04/2021] [Indexed: 11/30/2022] Open
Abstract
Apical membrane antigen 1 (AMA1) is a surface protein of Plasmodium sp. that plays a crucial role in forming moving junction (MJ) during the invasion of human red blood cells. The obligatory presence of AMA1 in the parasite lifecycle designates this protein as a potential vaccine candidate and an essential target for the development of novel peptide or protein therapeutics. However, due to multiple cysteine residues in the protein sequence, attaining the native fold with correct disulfide linkages during the refolding process after expression in bacteria has remained challenging for years. Although several approaches to obtain the refolded protein from bacterial expression have been reported previously, achieving high yield during refolding and proper functional validation of the expressed protein was lacking. We report here an improved method of refolding to obtain higher quantity of refolded protein. We have also validated the refolded protein's functional activity by evaluating the expressed AMA1 protein binding with a known inhibitory peptide, rhoptry neck protein 2 (RON2), using surface plasmon resonance (SPR) and isothermal titration calorimetry (ITC). A simple yet effective protocol for P. falciparum AMA1 protein expression from E. coli. Highly reproducible and scalable refolding protocol. The modified refolding method uses a step-wise dialysis technique. Functional validation of the refolded protein shown by binding with PfRON2 ectodomain using SPR and ITC.
Collapse
Affiliation(s)
- Anamika Biswas
- TIFR Centre for Interdisciplinary Sciences, Tata Institute of Fundamental Research Hyderabad, 36/p Gopanpally, Hyderabad, Telangana 500046, India
| | - Sreejith Raran-Kurussi
- TIFR Centre for Interdisciplinary Sciences, Tata Institute of Fundamental Research Hyderabad, 36/p Gopanpally, Hyderabad, Telangana 500046, India
| | - Akash Narayan
- TIFR Centre for Interdisciplinary Sciences, Tata Institute of Fundamental Research Hyderabad, 36/p Gopanpally, Hyderabad, Telangana 500046, India
| | - Abhisek Kar
- TIFR Centre for Interdisciplinary Sciences, Tata Institute of Fundamental Research Hyderabad, 36/p Gopanpally, Hyderabad, Telangana 500046, India
| | - Purna Chandra Mashurabad
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Gachibowli, Hyderabad, Telangana 500046, India
| | - Mrinal Kanti Bhattacharyya
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Gachibowli, Hyderabad, Telangana 500046, India
| | - Kalyaneswar Mandal
- TIFR Centre for Interdisciplinary Sciences, Tata Institute of Fundamental Research Hyderabad, 36/p Gopanpally, Hyderabad, Telangana 500046, India
- Corresponding author.
| |
Collapse
|
3
|
Global selective sweep of a highly inbred genome of the cattle parasite Neospora caninum. Proc Natl Acad Sci U S A 2019; 116:22764-22773. [PMID: 31636194 DOI: 10.1073/pnas.1913531116] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Neospora caninum, a cyst-forming apicomplexan parasite, is a leading cause of neuromuscular diseases in dogs as well as fetal abortion in cattle worldwide. The importance of the domestic and sylvatic life cycles of Neospora, and the role of vertical transmission in the expansion and transmission of infection in cattle, is not sufficiently understood. To elucidate the population genomics of Neospora, we genotyped 50 isolates collected worldwide from a wide range of hosts using 19 linked and unlinked genetic markers. Phylogenetic analysis and genetic distance indices resolved a single genotype of N. caninum Whole-genome sequencing of 7 isolates from 2 different continents identified high linkage disequilibrium, significant structural variation, but only limited polymorphism genome-wide, with only 5,766 biallelic single nucleotide polymorphisms (SNPs) total. Greater than half of these SNPs (∼3,000) clustered into 6 distinct haploblocks and each block possessed limited allelic diversity (with only 4 to 6 haplotypes resolved at each cluster). Importantly, the alleles at each haploblock had independently segregated across the strains sequenced, supporting a unisexual expansion model that is mosaic at 6 genomic blocks. Integrating seroprevalence data from African cattle, our data support a global selective sweep of a highly inbred livestock pathogen that originated within European dairy stock and expanded transcontinentally via unisexual mating and vertical transmission very recently, likely the result of human activities, including recurrent migration, domestication, and breed development of bovid and canid hosts within similar proximities.
Collapse
|
4
|
Identification of a peptide derived from a Bothrops moojeni metalloprotease with in vitro inhibitory action on the Plasmodium falciparum purine nucleoside phosphorylase enzyme (PfPNP). Biochimie 2019; 162:97-106. [PMID: 30978375 DOI: 10.1016/j.biochi.2019.04.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 04/07/2019] [Indexed: 11/22/2022]
Abstract
There is a growing need for research on new antimalarial agents against Plasmodium falciparum infection, especially in regards to planning molecular architecture for specific molecular targets of the parasite. Thus, a metalloprotease from Bothrops moojeni, known as BmooMPα-I, was explored in this study, through in silico assays, aiming at the development of a peptide generated from this molecule with potential inhibitory action on PfPNP, an enzyme necessary for the survival of the parasite. In order to isolate BmooMPα-I, cation exchange and reverse phase chromatographies were performed, followed by in vitro assays of antiparasitic activity against the W2 strain of P. falciparum. The interactions between BmooMPα-I and PfPNP were evaluated via docking, and the resulting peptide, described as Pep1 BM, was selected according to the BmooMPα-I region demonstrating the best interaction score with the target of interest. The values for the specific activities of the PfPNP reaction were measured using the inorganic phosphate substrate and MESG. The fraction corresponding to BmooMPα-I was identified as fraction 4 in the cation exchange chromatography step, due to proteolytic activity on casein and the presence of a major band at ≅ 23 kDa. BmooMPα-I was able to inhibit in vitro growth of W2 P. falciparum, with an IC50 value of 16.14 μg/mL. Virtual screening with Pep1 BM demonstrated two PfPNP target binding regions, with ΔG values at the interaction interface of -10.75 kcal/mol and -11.74 kcal/mol. A significant reduction in the enzymatic activity of PfPNP was observed in the presence of Pep 1 BM when compared to the assay in the absence of this possible inhibitor. BmooMPα-I showed activity in vitro against W2 P. falciparum. By means of in silico techniques, the Pep 1 BM was identified as having potential binding affinity to the catalytic site of PfPNP and of inhibiting its catalytic activity in vitro.
Collapse
|
5
|
Raturi V, Abhishek K, Jana S, Bag SS, Trivedi V. Virtual Screening, Molecular Modelling and Biochemical Studies to Exploit PF14_0660 as a Target to Identify Novel Anti-malarials. LETT DRUG DES DISCOV 2019. [DOI: 10.2174/1570180815666180727121200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
Malaria Parasite relies heavily on signal transduction pathways to control
growth, the progression of the life cycle and sustaining stress for its survival. Unlike kinases,
Plasmodium's phosphatome is one of the smallest and least explored for identifying drug target
for clinical intervention. PF14_0660 is a putative protein present on the chromosome 14 of
Plasmodium falciparum genome.
Methods:
Multiple sequence alignment of PF14_0660 with other known protein phosphatase indicate
the presence of phosphatase motif with specific residues essential for metal binding, catalysis
and providing structural stability. PF14_0660 is a mixed α/β type of protein with several β -sheet
and α-helix arranged to form βαβαβα sub-structure. The surface properties of PF14_0660 is conserved
with another phosphate of this family, but it profoundly diverges from the host protein tyrosine
phosphatase. PF14_0660 was cloned, over-expressed and protein is exhibiting phosphatase
activity in a dose-dependent manner. Docking of Heterocyclic compounds from chemical libraries
into the PF14_0660 active site found nice fitting of several candidate molecules.
Results:
Compound PPinh6, PPinh 7 and PPinh 5 are exhibiting antimalarial activity with an IC50
of 1.4 ± 0.2µM, 3.8 ± 0.3 µM and 9.4 ± 0.6µM respectively. Compound PPinh 6 and PPinh 7 are
inhibiting intracellular PF14_0660 phosphatase activity and killing parasite through the generation
of reactive oxygen species.
Conclusion:
Hence, a combination of molecular modelling, virtual screening and biochemical study
allowed us to explore the potentials of PF14_0660 as a drug target to design anti-malarials.
Collapse
Affiliation(s)
- Vimee Raturi
- Malaria Research Group, Department of Biosciences and Bioengineering, Indian Institute of Technology-Guwahati, Guwahati-781039, Assam, India
| | - Kumar Abhishek
- Malaria Research Group, Department of Biosciences and Bioengineering, Indian Institute of Technology-Guwahati, Guwahati-781039, Assam, India
| | - Subhashis Jana
- Department of Chemistry, Indian Institute of Technology-Guwahati, Guwahati- 781039, Assam, India
| | - Subhendu Sekhar Bag
- Department of Chemistry, Indian Institute of Technology-Guwahati, Guwahati- 781039, Assam, India
| | - Vishal Trivedi
- Malaria Research Group, Department of Biosciences and Bioengineering, Indian Institute of Technology-Guwahati, Guwahati-781039, Assam, India
| |
Collapse
|
6
|
A Review of Pharmacogenetics of Antimalarials and Associated Clinical Implications. Eur J Drug Metab Pharmacokinet 2018; 42:745-756. [PMID: 28070879 DOI: 10.1007/s13318-016-0399-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Genetic variability in drug-metabolizing enzymes and drug transporters is known to influence the pharmacokinetics of many drugs. Antimalarial drugs are a class of agents known to utilize metabolic and elimination pathways prone to genetic variation. This paper aims to review the genetic variants affecting antimalarial medications and discuss their clinical implications. Data were identified for the genes coding for the cytochrome P450 (CYP) enzymes: CYP2C8, CYP2C19, CYP2A6, CYP2D6, CYP2B6, and the P-glycoprotein drug transporter. Adverse effects of amodiaquine were more common in patients with decreased CYP2C8 metabolism. CYP2C19 variants influenced the metabolism of proguanil but no differences in efficacy outcomes were observed. Ultra-metabolizers of CYP2A6 showed increased incidence of adverse effects of artesunate (prodrug for active metabolite, dihydroartemisinin). In the presence of efavirenz, mutations in CYP2B6 influenced the number of patients achieving day-7 lumefantrine concentrations above accepted therapeutic cut-offs. Lumefantrine concentrations were also influenced by ABCB1 variants in the presence of nevirapine. The most critical pharmacogenetic consideration identified was the association of glucose-6-phosphate dehydrogenase deficiency with development of hemolytic anemia and decreased hemoglobin levels in patients treated with primaquine or a combination of chlorproguanil-dapsone-artesunate. These findings demonstrate a need for close monitoring of patients originating from populations where genetic variation in metabolizing enzymes is prevalent, so as to ensure that optimal clinical outcomes are achieved. Future studies should determine which populations are at greatest risk of potential treatment failures and/or adverse effects, which drugs are most susceptible to genetic variation in metabolizing enzymes, and the impact of genetic influence on the efficacy and safety of first-line treatment regimens.
Collapse
|
7
|
Abstract
Objectives: Malaria has been a major global health problem in recent times with increasing mortality. Current treatment methods include parasiticidal drugs and vaccinations. However, resistance among malarial parasites to the existing drugs has emerged as a significant area of concern in anti-malarial drug design. Researchers are now desperately looking for new targets to develop anti-malarials drug which is more target specific. Malarial parasites harbor a plastid-like organelle known as the ‘apicoplast’, which is thought to provide an exciting new outlook for the development of drugs to be used against the parasite. This review elaborates on the current state of development of novel compounds targeted againstemerging malaria parasites. Methods: The apicoplast, originates by an endosymbiotic process, contains a range of metabolic pathways and housekeeping processes that differ from the host body and thereby presents ideal strategies for anti-malarial drug therapy. Drugs are designed by targeting the unique mechanism of the apicoplasts genetic machinery. Several anabolic and catabolic processes, like fatty acid, isopenetyl diphosphate and heme synthess in this organelle, have also been targeted by drugs. Results: Apicoplasts offer exciting opportunities for the development of malarial treatment specific drugs have been found to act by disrupting this organelle’s function, which wouldimpede the survival of the parasite. Conclusion: Recent advanced drugs, their modes of action, and their advantages in the treatment of malaria by using apicoplasts as a target are discussed in this review which thought to be very useful in desigining anti-malarial drugs. Targetting the genetic machinery of apicoplast shows a great advantange regarding anti-malarial drug design. Critical knowledge of these new drugs would give a healthier understanding for deciphering the mechanism of action of anti-malarial drugs when targeting apicoplasts to overcome drug resistance.
Collapse
Affiliation(s)
- Avinaba Mukherjee
- Department of Pharmaceutical Technology, Natural Science Laboratory, Jadavpur University, Kolkata, India
| | | |
Collapse
|
8
|
Daniels R, Hamilton EJ, Durfee K, Ndiaye D, Wirth DF, Hartl DL, Volkman SK. Methods to Increase the Sensitivity of High Resolution Melting Single Nucleotide Polymorphism Genotyping in Malaria. J Vis Exp 2015:e52839. [PMID: 26575471 PMCID: PMC4692701 DOI: 10.3791/52839] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Despite decades of eradication efforts, malaria remains a global burden. Recent renewed interest in regional elimination and global eradication has been accompanied by increased genomic information about Plasmodium parasite species responsible for malaria, including characteristics of geographical populations as well as variations associated with reduced susceptibility to anti-malarial drugs. One common genetic variation, single-nucleotide polymorphisms (SNPs), offers attractive targets for parasite genotyping. These markers are useful not only for tracking drug resistance markers but also for tracking parasite populations using markers not under drug or other selective pressures. SNP genotyping methods offer the ability to track drug resistance as well as to fingerprint individual parasites for population surveillance, particularly in response to malaria control efforts in regions nearing elimination status. While informative SNPs have been identified that are agnostic to specific genotyping technologies, high-resolution melting (HRM) analysis is particularly suited to field-based studies. Compared to standard fluorescent-probe based methods that require individual SNPs in a single labeled probe and offer at best 10% sensitivity to detect SNPs in samples that contain multiple genomes (polygenomic), HRM offers 2-5% sensitivity. Modifications to HRM, such as blocked probes and asymmetric primer concentrations as well as optimization of amplification annealing temperatures to bias PCR towards amplification of the minor allele, further increase the sensitivity of HRM. While the sensitivity improvement depends on the specific assay, we have increased detection sensitivities to less than 1% of the minor allele. In regions approaching malaria eradication, early detection of emerging or imported drug resistance is essential for prompt response. Similarly, the ability to detect polygenomic infections and differentiate imported parasite types from cryptic local reservoirs can inform control programs. This manuscript describes modifications to high resolution melting technology that further increase its sensitivity to identify polygenomic infections in patient samples.
Collapse
Affiliation(s)
- Rachel Daniels
- Department of Organismic and Evolutionary Biology, Harvard University; Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health;
| | - Elizabeth J Hamilton
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health
| | - Katelyn Durfee
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health
| | - Daouda Ndiaye
- Faculty of Medicine and Pharmacy, Cheikh Anta Diop University
| | - Dyann F Wirth
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health; Institute of Infectious Diseases, Broad Institute
| | - Daniel L Hartl
- Department of Organismic and Evolutionary Biology, Harvard University
| | - Sarah K Volkman
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health; School of Nursing and Health Sciences, Simmons College
| |
Collapse
|
9
|
Maindola P, Jamal S, Grover A. Cheminformatics Based Machine Learning Models for AMA1-RON2 Abrogators for Inhibiting Plasmodium falciparum Erythrocyte Invasion. Mol Inform 2015; 34:655-664. [PMID: 27490966 DOI: 10.1002/minf.201400139] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 02/21/2015] [Indexed: 01/15/2023]
Abstract
Malaria remains a dreadful disease by putting every year about 3.4 billion people at risk and resulting into mortality of 627 thousand people worldwide. Existing therapies based upon Quinines and Artemisinin-based combination therapies have started showing resistance, pressing the need for search of anti-malarials with different mechanisms of action. In this respect erythrocyte invasion by Plasmodium is immensely crucial, as being obligate intracellular parasite it must invade host cells. This process is mediated by interaction between conserved Apical Membrane Antigen (AMA1) and Rhoptry Neck (RON2) protein, which is compulsory for successful invasion of erythrocyte by Plasmodium and manifestation of the disease Malaria. Here, using the physicochemical properties of the compounds available from a confirmatory high throughput screening, which were tested for their disruption capability of this crucial molecular interaction, we trained supervised classifiers and validated their robustness by various statistical parameters. Best model was used for screening new compounds from Traditional Chinese Medicine Database. Some of the best hits already find their use as anti-malarials and the model predicts that an essential part of their effectiveness is likely due to inhibition of AMA1-RON2 interaction. Pharmacophoric features have also been identified to ease further designing of possible leads in an effective way.
Collapse
Affiliation(s)
- Priyank Maindola
- School of Biotechnology, Jawaharlal Nehru University, New Delhi-110067, India phone/fax: +91-11-26738728; fax: +91-11-26702040
| | - Salma Jamal
- School of Biotechnology, Jawaharlal Nehru University, New Delhi-110067, India phone/fax: +91-11-26738728; fax: +91-11-26702040
| | - Abhinav Grover
- School of Biotechnology, Jawaharlal Nehru University, New Delhi-110067, India phone/fax: +91-11-26738728; fax: +91-11-26702040.
| |
Collapse
|
10
|
Modelling the effects of mass drug administration on the molecular epidemiology of schistosomes. ADVANCES IN PARASITOLOGY 2015; 87:293-327. [PMID: 25765198 DOI: 10.1016/bs.apar.2014.12.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
As national governments scale up mass drug administration (MDA) programs aimed to combat neglected tropical diseases (NTDs), novel selection pressures on these parasites increase. To understand how parasite populations are affected by MDA and how to maximize the success of control programmes, it is imperative for epidemiological, molecular and mathematical modelling approaches to be combined. Modelling of parasite population genetic and genomic structure, particularly of the NTDs, has been limited through the availability of only a few molecular markers to date. The landscape of infectious disease research is being dramatically reshaped by next-generation sequencing technologies and our understanding of how repeated selective pressures are shaping parasite populations is radically altering. Genomics can provide high-resolution data on parasite population structure, and identify how loci may contribute to key phenotypes such as virulence and/or drug resistance. We discuss the incorporation of genetic and genomic data, focussing on the recently sequenced Schistosoma spp., into novel mathematical transmission models to inform our understanding of the impact of MDA and other control methods. We summarize what is known to date, the models that exist and how population genetics has given us an understanding of the effects of MDA on the parasites. We consider how genetic and genomic data have the potential to shape future research, highlighting key areas where data are lacking, and how future molecular epidemiology knowledge can aid understanding of transmission dynamics and the effects of MDA, ultimately informing public health policy makers of the best interventions for NTDs.
Collapse
|
11
|
Awasthi G, Das A. Genetics of chloroquine-resistant malaria: a haplotypic view. Mem Inst Oswaldo Cruz 2015; 108:947-61. [PMID: 24402147 PMCID: PMC4005552 DOI: 10.1590/0074-0276130274] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Accepted: 09/26/2013] [Indexed: 02/05/2023] Open
Abstract
The development and rapid spread of chloroquine resistance (CQR) in
Plasmodium falciparum have triggered the identification of
several genetic target(s) in the P. falciparum genome. In
particular, mutations in the Pfcrt gene, specifically, K76T and
mutations in three other amino acids in the region adjoining K76 (residues 72, 74, 75
and 76), are considered to be highly related to CQR. These various mutations form
several different haplotypes and Pfcrt gene polymorphisms and the
global distribution of the different CQR- Pfcrt haplotypes in
endemic and non-endemic regions of P. falciparum malaria have been
the subject of extensive study. Despite the fact that the Pfcrt gene
is considered to be the primary CQR gene in P. falciparum , several
studies have suggested that this may not be the case. Furthermore, there is a poor
correlation between the evolutionary implications of the Pfcrt
haplotypes and the inferred migration of CQR P. falciparum based on
CQR epidemiological surveillance data. The present paper aims to clarify the existing
knowledge on the genetic basis of the different CQR- Pfcrt
haplotypes that are prevalent in worldwide populations based on the published
literature and to analyse the data to generate hypotheses on the genetics and
evolution of CQR malaria.
Collapse
|
12
|
Read AF, Huijben S. Evolutionary biology and the avoidance of antimicrobial resistance. Evol Appl 2015; 2:40-51. [PMID: 25567846 PMCID: PMC3352414 DOI: 10.1111/j.1752-4571.2008.00066.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2008] [Accepted: 12/11/2008] [Indexed: 11/29/2022] Open
Abstract
Evolutionary biologists have largely left the search for solutions to the drug resistance crisis to biomedical scientists, physicians, veterinarians and public health specialists. We believe this is because the vast majority of professional evolutionary biologists consider the evolutionary science of drug resistance to be conceptually uninteresting. Using malaria as case study, we argue that it is not. We review examples of evolutionary thinking that challenge various fallacies dominating antimalarial therapy, and discuss open problems that need evolutionary insight. These problems are unlikely to be resolved by biomedical scientists ungrounded in evolutionary biology. Involvement by evolutionary biologists in the science of drug resistance requires no intellectual compromises: the problems are as conceptually challenging as they are important.
Collapse
Affiliation(s)
- Andrew F Read
- Center for Infectious Disease Dynamics, Departments of Biology and Entomology, Pennsylvania State University, University Park PA, USA
| | - Silvie Huijben
- Center for Infectious Disease Dynamics, Departments of Biology and Entomology, Pennsylvania State University, University Park PA, USA ; School of Biological Sciences, University of Edinburgh West Mains Road, Edinburgh, UK
| |
Collapse
|
13
|
Kim Y, Escalante AA, Schneider KA. A population genetic model for the initial spread of partially resistant malaria parasites under anti-malarial combination therapy and weak intrahost competition. PLoS One 2014; 9:e101601. [PMID: 25007207 PMCID: PMC4090191 DOI: 10.1371/journal.pone.0101601] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Accepted: 06/09/2014] [Indexed: 11/18/2022] Open
Abstract
To develop public-health policies that extend the lifespan of affordable anti-malarial drugs as effective treatment options, it is necessary to understand the evolutionary processes leading to the origin and spread of mutations conferring drug resistance in malarial parasites. We built a population-genetic model for the emergence of resistance under combination drug therapy. Reproductive cycles of parasites are specified by their absolute fitness determined by clinical parameters, thus coupling the evolutionary-genetic with population-dynamic processes. Initial mutations confer only partial drug-resistance. Therefore, mutant parasites rarely survive combination therapy and within-host competition is very weak among parasites. The model focuses on the early phase of such unsuccessful recurrent mutations. This ends in the rare event of mutants enriching in an infected individual from which the successful spread of resistance over the entire population is initiated. By computer simulations, the waiting time until the establishment of resistant parasites is analysed. Resistance spreads quickly following the first appearance of a host infected predominantly by mutant parasites. This occurs either through a rare transmission of a resistant parasite to an uninfected host or through a rare failure of drugs in removing "transient" mutant alleles. The emergence of resistance is delayed with lower mutation rate, earlier treatment, higher metabolic cost of resistance, longer duration of high drug dose, and higher drug efficacy causing a stronger reduction in the sensitive and resistant parasites' fitnesses. Overall, contrary to other studies' proposition, the current model based on absolute fitness suggests that aggressive drug treatment delays the emergence of drug resistance.
Collapse
Affiliation(s)
- Yuseob Kim
- Department of Life Science and Division of EcoScience, Ewha Womans University, Seoul, South Korea
- * E-mail:
| | - Ananias A. Escalante
- School of Life Sciences and Center for Evolutionary Medicine and Informatics at the Biodesign Institute, Arizona State University, Tempe, Arizona, United States of America
| | | |
Collapse
|
14
|
Chauhan K, Pande V, Das A. DNA sequence polymorphisms of the pfmdr1 gene and association of mutations with the pfcrt gene in Indian Plasmodium falciparum isolates. INFECTION GENETICS AND EVOLUTION 2014; 26:213-22. [PMID: 24911283 DOI: 10.1016/j.meegid.2014.05.033] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2014] [Revised: 05/29/2014] [Accepted: 05/31/2014] [Indexed: 12/24/2022]
Abstract
Mutations in the Plasmodium falciparum multidrug resistance (pfmdr1) gene are known to provide compensatory fitness benefits to the chloroquine (CQ)-resistant malaria parasites and are often associated with specific mutations in the P. falciparum CQ resistant transporter (pfcrt) gene. Prevalence of the specific mutations in these two genes across different malaria endemic regions was mostly studies. However, reports on mutations in the pfmdr1 gene and their genetic associations with mutations in the pfcrt gene in Indian P. falciparum field isolates are scarce. We have sequenced a 560 bp region of pfmdr1 coding sequence in 64 P. falciparum isolates collected from different malaria endemic populations in India. Twenty out of these 64 isolates were laboratory cultured with known in vitro CQ sensitiveness (10 sensitive and 10 resistant). Three low frequency mutations (two non-synonymous and one synonymous) in the pfmdr1 gene were segregating in Indian isolates in addition to the predominant Y₈₆ and Y₁₈₄ ones, with high haplotype and nucleotide diversity in the field isolates in comparison to the cultured ones. No statistically significant genetic association between the mutations in the pfmdr1 and pfcrt gene could be detected; almost all observed associations were intragenic in nature. The results on the genetic diversity of the pfmdr1 gene were discussed in term of evolutionary perspectives in Indian P. falciparum, with possible future potential of gaining further insights on this gene in view of evolving malaria parasites resistant to artemisinin partner drugs.
Collapse
Affiliation(s)
- Kshipra Chauhan
- Evolutionary Genomics and Bioinformatics Laboratory, Division of Genomics and Bioinformatics, National Institute of Malaria Research, Sector 8, Dwarka, New Delhi 110077, India
| | - Veena Pande
- Department of Biotechnology, Kumaun University, Nainital 263001, Uttarakhand, India
| | - Aparup Das
- Evolutionary Genomics and Bioinformatics Laboratory, Division of Genomics and Bioinformatics, National Institute of Malaria Research, Sector 8, Dwarka, New Delhi 110077, India.
| |
Collapse
|
15
|
Niranjan Reddy B, Gupta B, Rao BP. Vector population manipulation for control of arboviruses--a novel prospect for India. PEST MANAGEMENT SCIENCE 2014; 70:517-523. [PMID: 24254373 DOI: 10.1002/ps.3676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Revised: 10/23/2013] [Accepted: 10/31/2013] [Indexed: 06/02/2023]
Abstract
India, the seventh largest country in the world, has diverse geographical and climatic regions with vast rural and peri-urban areas. Many are experiencing an escalation in the spread and intensity of numerous human diseases transmitted by insects. Classically, the management of these vector-borne diseases is underpinned by either chemical insecticides and/or environmental management targeted at the vector. However, these methods or their present implementation do not offer acceptable levels of control, and more effective and sustainable options are now available. Genetic strategies for the prevention of arbovirus transmission are most advanced for dengue and chikungunya, targeting their primary vector, Aedes aegypti. The national burden in terms of morbidity and mortality as a direct consequence of dengue virus in India is considered to be the largest worldwide, over 4 times that of any other country. Presently, new genetic technologies are undergoing field evaluation of their biosafety and efficacy in several countries. This paper discusses the merits of these approaches and argues for fair and transparent appraisal in India as a matter of urgency. Identification of any associated risks and their appropriate mitigation are fundamental to that process.
Collapse
Affiliation(s)
- Bp Niranjan Reddy
- School of Studies in Biotechnology, Jiwaji University, Gwalior, Madhya Pradesh, India
| | | | | |
Collapse
|
16
|
Srinivasan P, Yasgar A, Luci DK, Beatty WL, Hu X, Andersen J, Narum DL, Moch JK, Sun H, Haynes JD, Maloney DJ, Jadhav A, Simeonov A, Miller LH. Disrupting malaria parasite AMA1-RON2 interaction with a small molecule prevents erythrocyte invasion. Nat Commun 2014; 4:2261. [PMID: 23907321 PMCID: PMC3755449 DOI: 10.1038/ncomms3261] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 07/05/2013] [Indexed: 02/03/2023] Open
Abstract
Plasmodium falciparum resistance to artemisinin derivatives, the first-line antimalarial drug, drives the search for new classes of chemotherapeutic agents. Current discovery is primarily directed against the intracellular forms of the parasite. However, late schizont-infected red blood cells (RBCs) may still rupture and cause disease by sequestration; consequently targeting invasion may reduce disease severity. Merozoite invasion of RBCs requires interaction between two parasite proteins AMA1 and RON2. Here we identify the first inhibitor of this interaction that also blocks merozoite invasion in genetically distinct parasites by screening a library of over 21,000 compounds. We demonstrate that this inhibition is mediated by the small molecule binding to AMA1 and blocking the formation of AMA1-RON complex. Electron microscopy confirms that the inhibitor prevents junction formation, a critical step in invasion that results from AMA1-RON2 binding. This study uncovers a strategy that will allow for highly effective combination therapies alongside existing antimalarial drugs.
Collapse
Affiliation(s)
- Prakash Srinivasan
- Laboratory of Malaria and Vector Research, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20852, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Al-Hamidhi S, Mahdy MAK, Al-Hashami Z, Al-Farsi H, Al-mekhlafi AM, Idris MA, Beja-Pereira A, Babiker HA. Genetic diversity of Plasmodium falciparum and distribution of drug resistance haplotypes in Yemen. Malar J 2013; 12:244. [PMID: 23855834 PMCID: PMC3729657 DOI: 10.1186/1475-2875-12-244] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2013] [Accepted: 07/11/2013] [Indexed: 11/25/2022] Open
Abstract
Background Despite evident success of malaria control in many sites in the Arabian Peninsula, malaria remains endemic in a few spots, in Yemen and south-west of Saudi Arabia. In addition to local transmission, imported malaria sustains an extra source of parasites that can challenge the strengths of local control strategies. This study examined the genetic diversity of Plasmodium falciparum in Yemen and mutations of drug resistant genes, to elucidate parasite structure and distribution of drug resistance genotypes in the region. Methods Five polymorphic loci (MSP-2, Pfg377 and three microsatellites on chromosome 8) not involved in anti-malarial drug resistance, and four drug resistant genes (pfcrt, pfmdr1, dhfr and dhps) were genotyped in 108 P. falciparum isolates collected in three sites in Yemen: Dhamar, Hodeidah and Taiz. Results High diversity was seen in non-drug genes, pfg377 (He = 0.66), msp-2 (He = 0.80) and three microsatellites on chr 8, 7.7 kb (He = 0.88), 4.3 kb (He = 0.77) and 0.8 kb (He = 0.71). There was a high level of mixed-genotype infections (57%), with an average 1.8 genotypes per patient. No linkage disequilibrium was seen between drug resistant genes and the non-drug markers (p < 0.05). Genetic differentiation between populations was low (most pair-wise FST values <0.03), indicating extensive gene flow between the parasites in the three sites. There was a high prevalence of mutations in pfmdr1, pfcrt and dhfr; with four mutant pfmdr1 genotypes (NFCDD[57%], NFSND[21%], YFCDD[13%] and YFSND[8% ]), two mutant pfcrt genotypes (CVIET[89%] and SVMNT[4%]) and one mutant dhfr genotype (ICNI[53.7%]). However, no dhps mutations were detected. Conclusion The high diversity of P. falciparum in Yemen is indicative of a large parasite reservoir, which represents a challenge to control efforts. The presence of two distinct pfcrt genotype, CVIET and SVMNT, suggests that chloroquine resistance can possibly be related to a migratory path from Africa and Asia. The absence of the triple mutant dhfr genotype (IRN) and dhps mutations supports the use of artesunate + sulphadoxine-pyrimethamine as first-line therapy. However, the prevalent pfmdr1 genotype NFSND [21%] has previously been associated with tolerance/resistance response to artemisinin combination therapy (ACT). Regular surveys are, therefore, important to monitor spread of pfmdr1 and dhfr mutations and response to ACT.
Collapse
|
18
|
Abstract
INTRODUCTION The relict plastid, or apicoplast, is a characteristic feature of Plasmodium spp. and reflects the unusual evolutionary origins of these parasites. The essential role this organelle plays in the life of the parasite, and its unusual, non-mammalian metabolism, make the apicoplast an excellent drug target. AREAS COVERED This review focuses on the biological role of the apicoplast in the erythrocytic life cycle and what that reveals about existing drug targets. We also discuss the future of the apicoplast in the development of anti-malarials, emphasizing those pathways with greatest potential as a source of novel drug targets and emphasizing the need to understand in vitro drug responses to optimize eventual use of these drugs to treat malaria. EXPERT OPINION More than a decade of research on the apicoplast has confirmed the promise of this organelle as a source of drug targets. It is now possible to rationally assess the value of existing drugs and new drug targets, and to understand the role these drugs can play in the arsenal of anti-malarial treatments.
Collapse
Affiliation(s)
- Christopher D Goodman
- University of Melbourne, School of Botany, Professor's Walk, Parkville, Vic, 3010, Australia.
| | | |
Collapse
|
19
|
Abstract
Malaria is an important human disease and is the target of a global eradication campaign. New technological and informatics advancements in population genomics are being leveraged to identify genetic loci under selection in the malaria parasite and to find variants that are associated with key clinical phenotypes, such as drug resistance. This article provides a timely Review of how population-genetics-based strategies are being applied to Plasmodium falciparum both to identify genetic loci as key targets of interventions and to develop monitoring and surveillance tools that are crucial for the successful elimination and eradication of malaria.
Collapse
|
20
|
Kinga Modrzynska K, Creasey A, Loewe L, Cezard T, Trindade Borges S, Martinelli A, Rodrigues L, Cravo P, Blaxter M, Carter R, Hunt P. Quantitative genome re-sequencing defines multiple mutations conferring chloroquine resistance in rodent malaria. BMC Genomics 2012; 13:106. [PMID: 22435897 PMCID: PMC3362770 DOI: 10.1186/1471-2164-13-106] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Accepted: 03/21/2012] [Indexed: 01/18/2023] Open
Abstract
Background Drug resistance in the malaria parasite Plasmodium falciparum severely compromises the treatment and control of malaria. A knowledge of the critical mutations conferring resistance to particular drugs is important in understanding modes of drug action and mechanisms of resistances. They are required to design better therapies and limit drug resistance. A mutation in the gene (pfcrt) encoding a membrane transporter has been identified as a principal determinant of chloroquine resistance in P. falciparum, but we lack a full account of higher level chloroquine resistance. Furthermore, the determinants of resistance in the other major human malaria parasite, P. vivax, are not known. To address these questions, we investigated the genetic basis of chloroquine resistance in an isogenic lineage of rodent malaria parasite P. chabaudi in which high level resistance to chloroquine has been progressively selected under laboratory conditions. Results Loci containing the critical genes were mapped by Linkage Group Selection, using a genetic cross between the high-level chloroquine-resistant mutant and a genetically distinct sensitive strain. A novel high-resolution quantitative whole-genome re-sequencing approach was used to reveal three regions of selection on chr11, chr03 and chr02 that appear progressively at increasing drug doses on three chromosomes. Whole-genome sequencing of the chloroquine-resistant parent identified just four point mutations in different genes on these chromosomes. Three mutations are located at the foci of the selection valleys and are therefore predicted to confer different levels of chloroquine resistance. The critical mutation conferring the first level of chloroquine resistance is found in aat1, a putative aminoacid transporter. Conclusions Quantitative trait loci conferring selectable phenotypes, such as drug resistance, can be mapped directly using progressive genome-wide linkage group selection. Quantitative genome-wide short-read genome resequencing can be used to reveal these signatures of drug selection at high resolution. The identities of three genes (and mutations within them) conferring different levels of chloroquine resistance generate insights regarding the genetic architecture and mechanisms of resistance to chloroquine and other drugs. Importantly, their orthologues may now be evaluated for critical or accessory roles in chloroquine resistance in human malarias P. vivax and P. falciparum.
Collapse
|
21
|
Application of genomics to field investigations of malaria by the international centers of excellence for malaria research. Acta Trop 2012; 121:324-32. [PMID: 22182668 DOI: 10.1016/j.actatropica.2011.12.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2011] [Revised: 11/28/2011] [Accepted: 12/02/2011] [Indexed: 12/15/2022]
Abstract
Success of the global research agenda toward eradication of malaria will depend on development of new tools, including drugs, vaccines, insecticides and diagnostics. Genomic information, now available for the malaria parasites, their mosquito vectors, and human host, can be leveraged to both develop these tools and monitor their effectiveness. Although knowledge of genomic sequences for the malaria parasites, Plasmodium falciparum and Plasmodium vivax, have helped advance our understanding of malaria biology, simply knowing this sequence information has not yielded a plethora of new interventions to reduce the burden of malaria. Here we review and provide specific examples of how genomic information has increased our knowledge of parasite biology, focusing on P. falciparum malaria. We then discuss how population genetics can be applied toward the epidemiological and transmission-related goals outlined by the International Centers of Excellence for Malaria Research groups recently established by the National Institutes of Health. Finally, we propose genomics is a research area that can promote coordination and collaboration between various ICEMR groups, and that working together as a community can significantly advance the value of this information toward reduction of the global malaria burden.
Collapse
|
22
|
|
23
|
Yuan J, Cheng KCC, Johnson RL, Huang R, Pattaradilokrat S, Liu A, Guha R, Fidock D, Inglese J, Wellems TE, Austin CP, Su XZ. Chemical genomic profiling for antimalarial therapies, response signatures, and molecular targets. Science 2011; 333:724-9. [PMID: 21817045 PMCID: PMC3396183 DOI: 10.1126/science.1205216] [Citation(s) in RCA: 118] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Malaria remains a devastating disease largely because of widespread drug resistance. New drugs and a better understanding of the mechanisms of drug action and resistance are essential for fulfilling the promise of eradicating malaria. Using high-throughput chemical screening and genome-wide association analysis, we identified 32 highly active compounds and genetic loci associated with differential chemical phenotypes (DCPs), defined as greater than or equal to fivefold differences in half-maximum inhibitor concentration (IC(50)) between parasite lines. Chromosomal loci associated with 49 DCPs were confirmed by linkage analysis and tests of genetically modified parasites, including three genes that were linked to 96% of the DCPs. Drugs whose responses mapped to wild-type or mutant pfcrt alleles were tested in combination in vitro and in vivo, which yielded promising new leads for antimalarial treatments.
Collapse
Affiliation(s)
- Jing Yuan
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases
| | - Ken Chih-Chien Cheng
- NIH Chemical Genomics Center, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Ronald L. Johnson
- NIH Chemical Genomics Center, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Ruili Huang
- NIH Chemical Genomics Center, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Anna Liu
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases
| | - Rajarshi Guha
- NIH Chemical Genomics Center, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - David Fidock
- Departments of Microbiology and Immunology and of Medicine, Columbia University Medical Center, New York, New York, USA
| | - James Inglese
- NIH Chemical Genomics Center, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Thomas E. Wellems
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases
| | - Christopher P. Austin
- NIH Chemical Genomics Center, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Xin-zhuan Su
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases
| |
Collapse
|
24
|
Linkage maps from multiple genetic crosses and loci linked to growth-related virulent phenotype in Plasmodium yoelii. Proc Natl Acad Sci U S A 2011; 108:E374-82. [PMID: 21690382 DOI: 10.1073/pnas.1102261108] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Plasmodium yoelii is an excellent model for studying malaria pathogenesis that is often intractable to investigate using human parasites; however, genetic studies of the parasite have been hindered by lack of genome-wide linkage resources. Here, we performed 14 genetic crosses between three pairs of P. yoelii clones/subspecies, isolated 75 independent recombinant progeny from the crosses, and constructed a high-resolution linkage map for this parasite. Microsatellite genotypes from the progeny formed 14 linkage groups belonging to the 14 parasite chromosomes, allowing assignment of sequence contigs to chromosomes. Growth-related virulent phenotypes from 25 progeny of one of the crosses were significantly associated with a major locus on chromosome 13 and with two secondary loci on chromosomes 7 and 10. The chromosome 10 and 13 loci are both linked to day 5 parasitemia, and their effects on parasite growth rate are independent but additive. The locus on chromosome 7 is associated with day 10 parasitemia. The chromosome 13 locus spans ~220 kb of DNA containing 51 predicted genes, including the P. yoelii erythrocyte binding ligand, in which a C741Y substitution in the R6 domain is implicated in the change of growth rate. Similarly, the chromosome 10 locus spans ~234 kb with 71 candidate genes, containing a member of the 235-kDa rhoptry proteins (Py235) that can bind to the erythrocyte surface membrane. Atypical virulent phenotypes among the progeny were also observed. This study provides critical tools and information for genetic investigations of virulence and biology of P. yoelii.
Collapse
|
25
|
Limenitakis J, Soldati-Favre D. Functional genetics in Apicomplexa: potentials and limits. FEBS Lett 2011; 585:1579-88. [PMID: 21557944 DOI: 10.1016/j.febslet.2011.05.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2011] [Revised: 05/02/2011] [Accepted: 05/03/2011] [Indexed: 01/15/2023]
Abstract
The Apicomplexans are obligate intracellular protozoan parasites and the causative agents of severe diseases in humans and animals. Although complete genome sequences are available since many years and for several parasites, they are replete with putative genes of unassigned function. Forward and reverse genetic approaches are limited only to a few Apicomplexans that can either be propagated in vitro or in a convenient animal model. This review will compare and contrast the most recent strategies developed for the genetic manipulation of Plasmodium falciparum, Plasmodium berghei and Toxoplasma gondii that have taken advantage of the intrinsic features of their respective genomes. Efforts towards the improvement of the transfection efficiencies in malaria parasites, the development of approaches to study essential genes and the elaboration of high-throughput methods for the identification of gene function will be discussed.
Collapse
Affiliation(s)
- Julien Limenitakis
- Department of Microbiology and Molecular Medicine, CMU, University of Geneva, Geneva, Switzerland.
| | | |
Collapse
|
26
|
Identification and functional validation of the novel antimalarial resistance locus PF10_0355 in Plasmodium falciparum. PLoS Genet 2011; 7:e1001383. [PMID: 21533027 PMCID: PMC3080868 DOI: 10.1371/journal.pgen.1001383] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2010] [Accepted: 03/25/2011] [Indexed: 11/25/2022] Open
Abstract
The Plasmodium falciparum parasite's ability to adapt to
environmental pressures, such as the human immune system and antimalarial drugs,
makes malaria an enduring burden to public health. Understanding the genetic
basis of these adaptations is critical to intervening successfully against
malaria. To that end, we created a high-density genotyping array that assays
over 17,000 single nucleotide polymorphisms (∼1 SNP/kb), and applied it to
57 culture-adapted parasites from three continents. We characterized genome-wide
genetic diversity within and between populations and identified numerous loci
with signals of natural selection, suggesting their role in recent adaptation.
In addition, we performed a genome-wide association study (GWAS), searching for
loci correlated with resistance to thirteen antimalarials; we detected both
known and novel resistance loci, including a new halofantrine resistance locus,
PF10_0355. Through functional testing we demonstrated that
PF10_0355 overexpression decreases sensitivity to
halofantrine, mefloquine, and lumefantrine, but not to structurally unrelated
antimalarials, and that increased gene copy number mediates resistance. Our GWAS
and follow-on functional validation demonstrate the potential of genome-wide
studies to elucidate functionally important loci in the malaria parasite
genome. Malaria infection with the human pathogen Plasmodium falciparum
results in almost a million deaths each year, mostly in African children.
Efforts to eliminate malaria are underway, but the parasite is adept at eluding
both the human immune response and antimalarial treatments. Thus, it is
important to understand how the parasite becomes resistant to drugs and to
develop strategies to overcome resistance mechanisms. Toward this end, we used
population genetic strategies to identify genetic loci that contribute to
parasite adaptation and to identify candidate genes involved in drug resistance.
We examined over 17,000 genetic variants across the parasite genome in over 50
strains in which we also measured responses to many known antimalarial
compounds. We found a number of genetic loci showing signs of recent natural
selection and a number of loci potentially involved in modulating the
parasite's response to drugs. We further demonstrated that one of the novel
candidate genes (PF10_0355) modulates resistance to the
antimalarial compounds halofantrine, mefloquine, and lumefantrine. Overall, this
study confirms that we can use genome-wide approaches to identify clinically
relevant genes and demonstrates through functional testing that at least one of
these candidate genes is indeed involved in antimalarial drug resistance.
Collapse
|
27
|
Tan JC, Miller BA, Tan A, Patel JJ, Cheeseman IH, Anderson TJC, Manske M, Maslen G, Kwiatkowski DP, Ferdig MT. An optimized microarray platform for assaying genomic variation in Plasmodium falciparum field populations. Genome Biol 2011; 12:R35. [PMID: 21477297 PMCID: PMC3218861 DOI: 10.1186/gb-2011-12-4-r35] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2011] [Revised: 03/04/2011] [Accepted: 04/08/2011] [Indexed: 11/13/2022] Open
Abstract
We present an optimized probe design for copy number variation (CNV) and SNP genotyping in the Plasmodium falciparum genome. We demonstrate that variable length and isothermal probes are superior to static length probes. We show that sample preparation and hybridization conditions mitigate the effects of host DNA contamination in field samples. The microarray and workflow presented can be used to identify CNVs and SNPs with 95% accuracy in a single hybridization, in field samples containing up to 92% human DNA contamination.
Collapse
Affiliation(s)
- John C Tan
- The Eck Institute for Global Health, University of Notre Dame, 100 Galvin Life Sciences, Notre Dame, IN 46556, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
A wide range of drug transport studies using intact infected red blood cells, isolated malarial parasites, heterologous expression systems, and purified protein, combined with elegant genetic experiments, have suggested that chloroquine transport by the Plasmodium falciparum chloroquine resistance transporter (PfCRT) is a key aspect of the molecular mechanism of quinoline antimalarial drug resistance. However, many questions remain. This short review summarizes data that have led to drug channel versus drug pump hypotheses for PfCRT and suggests ways in which recent contrasting interpretations might be reconciled.
Collapse
Affiliation(s)
- Paul D Roepe
- Department of Chemistry, Department of Biochemistry and Cellular and Molecular Biology, Georgetown University, Washington, DC 20057, United States.
| |
Collapse
|
29
|
Müller IB, Hyde JE. Antimalarial drugs: modes of action and mechanisms of parasite resistance. Future Microbiol 2010; 5:1857-73. [DOI: 10.2217/fmb.10.136] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Malaria represents one of the most serious threats to human health worldwide, and preventing and curing this parasitic disease still depends predominantly on the administration of a small number of drugs whose efficacy is continually threatened and eroded by the emergence of drug-resistant parasite populations. This has an enormous impact on the mortality and morbidity resulting from malaria infection, especially in sub-Saharan Africa, where the lethal human parasite species Plasmodium falciparum accounts for approximately 90% of deaths recorded globally. Successful treatment of uncomplicated malaria is now highly dependent on artemisinin-based combination therapies. However, the first cases of artemisinin-resistant field isolates have been reported recently and potential replacement antimalarials are only in the developmental stages. Here, we summarize recent progress in tackling the problem of parasite resistance and discuss the underlying molecular mechanisms that confer resistance to current antimalarial agents as far as they are known, understanding of which should assist in the rational development of new drugs and the more effective deployment of older ones.
Collapse
Affiliation(s)
- Ingrid B Müller
- Department of Biochemistry, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - John E Hyde
- Manchester Interdisciplinary Biocentre, Faculty of Life Sciences, University of Manchester, 131 Princess Street, Manchester, M1 7DN, UK
| |
Collapse
|
30
|
Fandeur T, Abani M. [Subsidized artemisinin based combination treatments for Africa]. Med Mal Infect 2010; 40:611-6. [PMID: 21051164 DOI: 10.1016/j.medmal.2010.07.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2010] [Revised: 06/20/2010] [Accepted: 07/20/2010] [Indexed: 11/19/2022]
Abstract
Malaria-associated mortality and morbidity have increased in recent decades, with the worldwide spread of chloroquine and sulfadoxine-pyrimethamine resistant parasites. Artemisinin-based combination therapies (ACTs) have been proposed as an alternative to conventional antimalarial drugs. ACTs are effective against multidrug-resistant infections, work quickly, are safe and well tolerated, and seem to decrease transmission by inactivating gametocytes. The affordable medicines facility-malaria (AMFm) - an initiative aiming at increasing the availability of affordable ACTs through public and private practice - is trying to accelerate the large-scale use of ACT worldwide. This began with an initial pilot phase in a selected group of African countries. However, the epidemiology of malaria, the economic context, and healthcare infrastructure of African countries differ considerably from those prevailing in Asia, where ACTs were first implemented in the 1990s. ACT implementation in Africa must therefore be accompanied by control and operational measures to maintain the efficacy of ACT and to protect patients against misuse. We discuss the expected benefits of the AMFm initiative in Africa and stress the importance of dealing with operational issues before implementation in the field, focusing particularly on drug resistance.
Collapse
Affiliation(s)
- T Fandeur
- Unité de parasitologie, centre de recherche médicale et sanitaire, 634, boulevard de la Nation, YN034, Niamey, Niger.
| | | |
Collapse
|
31
|
Rottmann M, McNamara C, Yeung BKS, Lee MCS, Zou B, Russell B, Seitz P, Plouffe DM, Dharia NV, Tan J, Cohen SB, Spencer KR, González-Páez GE, Lakshminarayana SB, Goh A, Suwanarusk R, Jegla T, Schmitt EK, Beck HP, Brun R, Nosten F, Renia L, Dartois V, Keller TH, Fidock DA, Winzeler EA, Diagana TT. Spiroindolones, a potent compound class for the treatment of malaria. Science 2010; 329:1175-80. [PMID: 20813948 PMCID: PMC3050001 DOI: 10.1126/science.1193225] [Citation(s) in RCA: 939] [Impact Index Per Article: 62.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Recent reports of increased tolerance to artemisinin derivatives--the most recently adopted class of antimalarials--have prompted a need for new treatments. The spirotetrahydro-beta-carbolines, or spiroindolones, are potent drugs that kill the blood stages of Plasmodium falciparum and Plasmodium vivax clinical isolates at low nanomolar concentration. Spiroindolones rapidly inhibit protein synthesis in P. falciparum, an effect that is ablated in parasites bearing nonsynonymous mutations in the gene encoding the P-type cation-transporter ATPase4 (PfATP4). The optimized spiroindolone NITD609 shows pharmacokinetic properties compatible with once-daily oral dosing and has single-dose efficacy in a rodent malaria model.
Collapse
Affiliation(s)
- Matthias Rottmann
- Swiss Tropical and Public Health Institute, Parasite Chemotherapy, CH-4002 Basel, Switzerland
- University of Basel, CH-4003 Basel, Switzerland
| | - Case McNamara
- Genomics Institute of the Novartis Research Foundation, San Diego, CA 92121, USA
| | | | - Marcus C. S. Lee
- Department of Microbiology and Immunology, Columbia University Medical Center, NY 10032, USA
| | - Bin Zou
- Novartis Institute for Tropical Diseases, 138670 Singapore
| | - Bruce Russell
- Laboratory of Malaria Immunobiology, Singapore Immunology Network, Agency for Science Technology and Research (ASTAR), Biopolis, Singapore
| | - Patrick Seitz
- Swiss Tropical and Public Health Institute, Parasite Chemotherapy, CH-4002 Basel, Switzerland
- University of Basel, CH-4003 Basel, Switzerland
| | - David M. Plouffe
- Genomics Institute of the Novartis Research Foundation, San Diego, CA 92121, USA
| | - Neekesh V. Dharia
- Department of Cell Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jocelyn Tan
- Novartis Institute for Tropical Diseases, 138670 Singapore
| | - Steven B. Cohen
- Genomics Institute of the Novartis Research Foundation, San Diego, CA 92121, USA
| | - Kathryn R. Spencer
- Department of Cell Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | | | - Anne Goh
- Novartis Institute for Tropical Diseases, 138670 Singapore
| | - Rossarin Suwanarusk
- Laboratory of Malaria Immunobiology, Singapore Immunology Network, Agency for Science Technology and Research (ASTAR), Biopolis, Singapore
| | - Tim Jegla
- Department of Cell Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Esther K. Schmitt
- Natural Products Unit, Novartis Pharma AG, CH-4002 Basel, Switzerland
| | - Hans-Peter Beck
- Swiss Tropical and Public Health Institute, Parasite Chemotherapy, CH-4002 Basel, Switzerland
- University of Basel, CH-4003 Basel, Switzerland
| | - Reto Brun
- Swiss Tropical and Public Health Institute, Parasite Chemotherapy, CH-4002 Basel, Switzerland
- University of Basel, CH-4003 Basel, Switzerland
| | - Francois Nosten
- Shoklo Malaria Research Unit, Mae Sot, Tak 63110, Thailand
- Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine, Nuffield Department of Clinical Medicine, University of Oxford, OX3 7LJ, UK
| | - Laurent Renia
- Laboratory of Malaria Immunobiology, Singapore Immunology Network, Agency for Science Technology and Research (ASTAR), Biopolis, Singapore
| | | | | | - David A. Fidock
- Department of Microbiology and Immunology, Columbia University Medical Center, NY 10032, USA
- Department of Medicine (Division of Infectious Diseases), Columbia University Medical Center, NY 10032, USA
| | - Elizabeth A. Winzeler
- Genomics Institute of the Novartis Research Foundation, San Diego, CA 92121, USA
- Department of Cell Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | |
Collapse
|
32
|
|
33
|
Casano G, Dumètre A, Pannecouque C, Hutter S, Azas N, Robin M. Anti-HIV and antiplasmodial activity of original flavonoid derivatives. Bioorg Med Chem 2010; 18:6012-23. [PMID: 20638854 DOI: 10.1016/j.bmc.2010.06.067] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2010] [Revised: 06/10/2010] [Accepted: 06/21/2010] [Indexed: 10/19/2022]
Abstract
In our search for potent anti-HIV and antiplasmodial agents, novel series of flavonoid derivatives and their chalcone intermediates were synthesized and evaluated for inhibition of HIV multiplication and antiproliferative activity on Plasmodium falciparum parasites. Chalcones exhibited a more selective antiplasmodial activity than flavonoids. Methoxyflavone 7e was the only one compound active in both P. falciparum and HIV-1 whereas aminomethoxyflavones showed activity against HIV-2. Para substitution on the B ring seemed to increase HIV-2 potency.
Collapse
Affiliation(s)
- Gilles Casano
- Aix-Marseille Université, Institut des Sciences Moléculaires de Marseille, iSm2-UMR CNRS, Centre Saint Jérôme, Marseille Cedex 20, France
| | | | | | | | | | | |
Collapse
|
34
|
Amina K, Giuliana G, Prato M. From control to eradication of malaria: the end of being stuck in second gear? ASIAN PAC J TROP MED 2010. [DOI: 10.1016/s1995-7645(10)60101-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
|
35
|
Chiyaka C, Mukandavire Z, Das P, Nyabadza F, Hove-Musekwa SD, Mwambi H. Theoretical analysis of mixed Plasmodium malariae and Plasmodium falciparum infections with partial cross-immunity. J Theor Biol 2010; 263:169-78. [DOI: 10.1016/j.jtbi.2009.10.032] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2009] [Revised: 09/03/2009] [Accepted: 10/28/2009] [Indexed: 11/25/2022]
|
36
|
Pierson JT, Dumètre A, Hutter S, Delmas F, Laget M, Finet JP, Azas N, Combes S. Synthesis and antiprotozoal activity of 4-arylcoumarins. Eur J Med Chem 2010; 45:864-9. [DOI: 10.1016/j.ejmech.2009.10.022] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2009] [Revised: 10/09/2009] [Accepted: 10/15/2009] [Indexed: 11/28/2022]
|
37
|
Mu J, Myers RA, Jiang H, Liu S, Ricklefs S, Waisberg M, Chotivanich K, Wilairatana P, Krudsood S, White NJ, Udomsangpetch R, Cui L, Ho M, Ou F, Li H, Song J, Li G, Wang X, Seila S, Sokunthea S, Socheat D, Sturdevant DE, Porcella SF, Fairhurst RM, Wellems TE, Awadalla P, Su XZ. Plasmodium falciparum genome-wide scans for positive selection, recombination hot spots and resistance to antimalarial drugs. Nat Genet 2010; 42:268-71. [PMID: 20101240 PMCID: PMC2828519 DOI: 10.1038/ng.528] [Citation(s) in RCA: 166] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2009] [Accepted: 12/04/2009] [Indexed: 01/17/2023]
Abstract
Antimalarial drugs impose strong pressure on Plasmodium falciparum parasites and leave signatures of selection in the parasite genome 1,2. Search for signals of selection may lead to genes encoding drug or immune targets 3. The lack of high-throughput genotyping methods, inadequate knowledge of parasite population history, and time-consuming adaptations of parasites to in vitro culture have hampered genome-wide association studies (GWAS) of parasite traits. Here we report genotyping of DNA from 189 culture-adapted P. falciparum parasites using a custom-built array with thousands of single nucleotide polymorphisms (SNPs). Population structure, variation in recombination rate, and loci under recent positive selection were detected. Parasite half maximum inhibitory concentrations (IC50) to seven antimalarial drugs were obtained and used in GWAS to identify genes associated with drug responses. The SNP array and genome-wide parameters provide valuable tools and information for new advances in P. falciparum genetics.
Collapse
Affiliation(s)
- Jianbing Mu
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, Maryland, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Escalante AA, Smith DL, Kim Y. The dynamics of mutations associated with anti-malarial drug resistance in Plasmodium falciparum. Trends Parasitol 2009; 25:557-63. [PMID: 19864183 DOI: 10.1016/j.pt.2009.09.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2009] [Revised: 07/22/2009] [Accepted: 09/10/2009] [Indexed: 10/20/2022]
Abstract
The evolution of resistance in Plasmodium falciparum against safe and affordable drugs such as chloroquine (CQ) and sulfadoxine-pyrimethamine (SP) is a major global health threat. Investigating the dynamics of resistance against these antimalarial drugs will lead to approaches for addressing the problem of resistance in malarial parasites that are solidly based in evolutionary genetics and population biology. In this article, we discuss current developments in population biology modeling and evolutionary genetics. Despite great advancements achieved in the past decade, understanding the complex dynamics of mutations conferring drug resistance in P. falciparum requires approaches that consider the parasite population structure among other demographic processes.
Collapse
Affiliation(s)
- Ananias A Escalante
- School of Life Sciences, Arizona State University, PO Box 874501, Tempe, AZ 85287-4501, USA.
| | | | | |
Collapse
|
39
|
Hyde JE. Mapping the origins and spread of antifolate-resistant malaria parasites. Future Microbiol 2009; 4:953-8. [PMID: 19824787 DOI: 10.2217/fmb.09.66] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Evaluation of: Pearce RJ, Pota H, Evehe MSB et al.: Multiple origins and regional dispersal of resistant dhps in African Plasmodium falciparum malaria. PLoS Med. 6(4), e1000055 (2009). Widespread resistance to current antimalarial drugs is a major factor in the extremely high levels of mortality and disabling illness that still prevail in many developing countries. It is important to understand how frequently resistant malaria parasite strains arise and their patterns of propagation and dispersal across borders and continents. By studying the DNA sequences of both the gene encoding the drug target and its flanking regions, it is possible to collect and map such data, providing a considerable asset in devising and evaluating future strategies of drug use and deployment. In this article, Pearce et al. analyze a large number of parasite samples collected over a decade from countries across Africa, allowing them to present for the first time a detailed picture of the origins and relatively recent spread of resistance to sulfa-drugs, key components of antifolate drug combinations that have been used extensively as part of the antimalarial armory.
Collapse
Affiliation(s)
- John E Hyde
- Manchester Interdisciplinary Biocentre, Faculty of Life Sciences, University of Manchester, Manchester M1 7ND, UK.
| |
Collapse
|
40
|
Genetic mapping of targets mediating differential chemical phenotypes in Plasmodium falciparum. Nat Chem Biol 2009; 5:765-71. [PMID: 19734910 PMCID: PMC2784992 DOI: 10.1038/nchembio.215] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2009] [Accepted: 07/09/2009] [Indexed: 11/08/2022]
|
41
|
Menegon M, Pearce RJ, Inojosa WO, Pisani V, Abel PM, Matondo A, Bisoffi Z, Majori G, Ord R, Warhurst DC, Roper C, Severini C. Monitoring for multidrug-resistant Plasmodium falciparum isolates and analysis of pyrimethamine resistance evolution in Uige province, Angola. Trop Med Int Health 2009; 14:1251-7. [PMID: 19708897 DOI: 10.1111/j.1365-3156.2009.02369.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
OBJECTIVES To assess the extent of drug resistance in Uige through molecular genetic analysis and to test whether the dhfr triple mutant alleles present in Angola are of southeast Asian origin. METHODS Seventy-one samples of blood from children admitted to the Pediatric Emergency Unit of Uige Provincial Hospital in 2004 were screened for resistance mutations at pfcrt, pfmdr1, pfdhfr, pfdhps and pfATPase6. RESULTS Mutations in pfcrt (codon76), pfmdr1 (codon86), pfdhfr (codons 51, 59, 108) and pfdhps (codons 436, 437) were common. Among the 66 isolates for which we were able to determine complete genetic information 13.7% carried all seven of these mutations. Flanking microsatellite analysis revealed the triple mutant pfdhfr was derived from the southeast Asian lineage, while the N51I+S108N double mutant pfdhfr alleles are a local origin. pfATPase6 mutations were rare and S769N was not found. CONCLUSION The parasite population of Uige Angola has high frequency mutations in pfcrt, dhfr and dhps associated with resistance to chloroquine and sulphadoxine pyrimethamine, reflecting past reliance on these two drugs which were the mainstay of treatment until recently. Our findings show that drug resistance in Uige has occurred through a combination of local drug pressure and the regional and international dispersal of resistance mutant alleles.
Collapse
Affiliation(s)
- Michela Menegon
- Department of Infectious, Parasitic and Immunomediated Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Drug resistance mechanisms in helminths: is it survival of the fittest? Trends Parasitol 2009; 25:328-35. [DOI: 10.1016/j.pt.2009.04.004] [Citation(s) in RCA: 114] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2008] [Revised: 03/25/2009] [Accepted: 04/02/2009] [Indexed: 11/20/2022]
|
43
|
Ollomo B, Durand P, Prugnolle F, Douzery E, Arnathau C, Nkoghe D, Leroy E, Renaud F. A new malaria agent in African hominids. PLoS Pathog 2009; 5:e1000446. [PMID: 19478877 PMCID: PMC2680981 DOI: 10.1371/journal.ppat.1000446] [Citation(s) in RCA: 114] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2009] [Accepted: 04/25/2009] [Indexed: 11/24/2022] Open
Abstract
Plasmodium falciparum is the major human malaria agent responsible for 200 to 300 million infections and one to three million deaths annually, mainly among African infants. The origin and evolution of this pathogen within the human lineage is still unresolved. A single species, P. reichenowi, which infects chimpanzees, is known to be a close sister lineage of P. falciparum. Here we report the discovery of a new Plasmodium species infecting Hominids. This new species has been isolated in two chimpanzees (Pan troglodytes) kept as pets by villagers in Gabon (Africa). Analysis of its complete mitochondrial genome (5529 nucleotides including Cyt b, Cox I and Cox III genes) reveals an older divergence of this lineage from the clade that includes P. falciparum and P. reichenowi (∼21±9 Myrs ago using Bayesian methods and considering that the divergence between P. falciparum and P. reichenowi occurred 4 to 7 million years ago as generally considered in the literature). This time frame would be congruent with the radiation of hominoids, suggesting that this Plasmodium lineage might have been present in early hominoids and that they may both have experienced a simultaneous diversification. Investigation of the nuclear genome of this new species will further the understanding of the genetic adaptations of P. falciparum to humans. The risk of transfer and emergence of this new species in humans must be now seriously considered given that it was found in two chimpanzees living in contact with humans and its close relatedness to the most virulent agent of malaria. In 2002, the publication of the genome of Plasmodium falciparum, the most malignant agent of malaria, generated hopes in the fight against this deadly disease by the opportunities it offered to discover new drug targets. Since then results have not lived up to the expectations. The development of comparative genomics to further understanding of P. falciparum has indeed been hindered by a lack of knowledge of closely related species' genomes. Only one species, P. reichenowi, infecting chimpanzees, was hitherto known as a sister lineage of P. falciparum. Here we describe a new Plasmodium species infecting chimpanzees in Africa. Based on its whole mitochondrial genome, we demonstrate that this species is a relative of P. falciparum and P. reichenowi. The analysis of its genome should thus offer the opportunity to explore P. falciparum specific adaptations to humans. Our results bring new elements to the debate surrounding the origin of this lineage. They suggest that it may have been present in early hominoids and may have experienced a radiation congruent with that of its hosts. Our discovery highlights the paucity of our knowledge on the richness of Plasmodium species infecting primates and calls for more research in this direction.
Collapse
Affiliation(s)
- Benjamin Ollomo
- Unité des Maladies Virales Émergentes, Centre International de Recherches Médicales de Franceville, Franceville, Gabon
| | - Patrick Durand
- Laboratoire Génétique et Evolution des Maladies Infectieuses, UMR 2724 CNRS-IRD, IRD Montpellier, Montpellier, France
| | - Franck Prugnolle
- Laboratoire Génétique et Evolution des Maladies Infectieuses, UMR 2724 CNRS-IRD, IRD Montpellier, Montpellier, France
- * E-mail: (FP); (FR)
| | - Emmanuel Douzery
- Laboratoire de Paléontologie, Phylogénie & Paléobiologie, Institut des Sciences de l'Evolution (UMR 5554 CNRS), Université Montpellier II, Place E. Bataillon, Montpellier, France
| | - Céline Arnathau
- Laboratoire Génétique et Evolution des Maladies Infectieuses, UMR 2724 CNRS-IRD, IRD Montpellier, Montpellier, France
| | - Dieudonné Nkoghe
- Unité des Maladies Virales Émergentes, Centre International de Recherches Médicales de Franceville, Franceville, Gabon
| | - Eric Leroy
- Unité des Maladies Virales Émergentes, Centre International de Recherches Médicales de Franceville, Franceville, Gabon
- Unité Emergence des Pathologies Virales, UMR 190 IRD-Université de la Méditerranée, CIRMF, Franceville, Gabon
| | - François Renaud
- Laboratoire Génétique et Evolution des Maladies Infectieuses, UMR 2724 CNRS-IRD, IRD Montpellier, Montpellier, France
- * E-mail: (FP); (FR)
| |
Collapse
|
44
|
Su XZ, Jiang H, Yi M, Mu J, Stephens RM. Large-scale genotyping and genetic mapping in Plasmodium parasites. THE KOREAN JOURNAL OF PARASITOLOGY 2009; 47:83-91. [PMID: 19488413 DOI: 10.3347/kjp.2009.47.2.83] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2008] [Accepted: 04/03/2009] [Indexed: 11/23/2022]
Abstract
The completion of many malaria parasite genomes provides great opportunities for genomewide characterization of gene expression and high-throughput genotyping. Substantial progress in malaria genomics and genotyping has been made recently, particularly the development of various microarray platforms for large-scale characterization of the Plasmodium falciparum genome. Microarray has been used for gene expression analysis, detection of single nucleotide polymorphism (SNP) and copy number variation (CNV), characterization of chromatin modifications, and other applications. Here we discuss some recent advances in genetic mapping and genomic studies of malaria parasites, focusing on the use of high-throughput arrays for the detection of SNP and CNV in the P. falciparum genome. Strategies for genetic mapping of malaria traits are also discussed.
Collapse
Affiliation(s)
- Xin-Zhuan Su
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA.
| | | | | | | | | |
Collapse
|
45
|
Sauvage V, Aubert D, Escotte-Binet S, Villena I. The role of ATP-binding cassette (ABC) proteins in protozoan parasites. Mol Biochem Parasitol 2009; 167:81-94. [PMID: 19464325 DOI: 10.1016/j.molbiopara.2009.05.005] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2009] [Revised: 05/11/2009] [Accepted: 05/12/2009] [Indexed: 10/20/2022]
Abstract
The ATP-binding cassette (ABC) superfamily is one of the largest protein families with representatives in all kingdoms of life. Members of this superfamily are involved in a wide variety of transport processes with substrates ranging from small ions to relatively large polypeptides and polysaccharides, but also in cellular processes such as DNA repair, translation or regulation of gene expression. For many years, the role of ABC proteins was mainly investigated for their implication in drug resistance. However, recent studies focused rather on their physiological functions for the parasite. In this review, we present an overview of ABC proteins in major protozoan parasites including Leishmania, Trypanosoma, Plasmodium, Toxoplasma, Cryptosporidium and Entamoeba species. We will also discuss the role of characterized ABC transporters in the biology of the parasite and in drug resistance.
Collapse
Affiliation(s)
- Virginie Sauvage
- Laboratoire de Parasitologie-Mycologie, EA 3800, IFR 53, UFR Médecine, Université de Reims Champagne-Ardenne, 51 rue Cognacq-Jay, 51095 Reims Cedex, France
| | | | | | | |
Collapse
|