Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Bioorg Med Chem Lett ; 54: 128442, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34763083

ABSTRACT

The FDA approved drug Dronabinol was identified in a previous study applying virtual screening using the haemozoin crystal as a target against malaria parasites. The active ingredient of dronabinol is synthetic tetrahydrocannabinol (THC), which is one of the major cannabinoids from Cannabis sativa. Traditional use of cannabis for malaria fever was reported in the world's oldest pharmacopoeia, dating to around 5000 years ago. In this research we report that THC inhibits ß-haematin (synthetic haemozoin) and malaria parasite growth. Due the psychoactivity of THC, CBD, the other major naturally occurring cannabinoid that lacks the off-target psychoactive effects of THC, was also tested and inhibited ß-haematin but showed only a mild antimalarial activity. To evaluate whether THC inhibit haemozoin formation, we performed a cellular haem fractionation assay that indicated that is not the likely mechanism of action. For the first time, the cannabinoid chemical structure is raised as a new chemical class to be further studied for malaria treatment, aiming to overcome the undesirable psychoactive effects of THC and optimize the antimalarial effects.


Subject(s)
Antimalarials/pharmacology , Dronabinol/pharmacology , Malaria/drug therapy , Plasmodium falciparum/drug effects , Antimalarials/chemistry , Cannabis/chemistry , Dose-Response Relationship, Drug , Dronabinol/chemistry , HL-60 Cells , Hemeproteins/antagonists & inhibitors , Humans , Molecular Structure , Parasitic Sensitivity Tests , Structure-Activity Relationship
2.
Chembiochem ; 22(9): 1568-1572, 2021 05 04.
Article in English | MEDLINE | ID: mdl-33453069

ABSTRACT

A fluorescent analogue of a previously synthesised N,N-chelated IrIII complex was prepared by coordination of the organic ligand to an extrinsic bis(2-phenylpyridine)iridium(III) fluorophore. This cyclometallated IrIII complex in itself displays good, micromolar activity against the chloroquine-sensitive NF54 strain of Plasmodium falciparum. Live-cell confocal microscopy found negligible localisation of the fluorescent complex within the digestive vacuole of the parasite. This eliminated the haem detoxification pathway as a potential mechanism of action. Similarly, no localisation of the complex within the parasitic nucleus was found, thus suggesting that this complex probably does not interfere with the DNA replication process. A substantial saturation of fluorescence from the complex was found near phospholipid structures such as the plasma and nuclear membranes but not in neutral lipid bodies. This indicates that an association with these membranes, or organelles such as the endoplasmic reticulum or branched mitochondrion, could be essential to the efficacies of these types of antimalarial compounds.


Subject(s)
Antimalarials/pharmacology , Coordination Complexes/chemistry , Iridium/chemistry , Plasmodium falciparum/drug effects , Quinolines/chemistry , Antimalarials/chemistry , Coordination Complexes/pharmacology , Erythrocytes/cytology , Erythrocytes/parasitology , Erythrocytes/pathology , Fluorescent Dyes/chemistry , Humans , Microscopy, Confocal
3.
ACS Infect Dis ; 7(2): 362-376, 2021 02 12.
Article in English | MEDLINE | ID: mdl-33430579

ABSTRACT

A diverse series of hemozoin-inhibiting quinolines, benzamides, triarylimidazoles, quinazolines, benzimidazoles, benzoxazoles, and benzothiazoles have been found to lead to exchangeable heme levels in cultured Plasmodium falciparum (NF54) that ranged over an order of magnitude at the IC50. Surprisingly, less active compounds often exhibited higher levels of exchangeable heme than more active ones. Quantities of intracellular inhibitor measured using the inoculum effect exhibited a linear correlation with exchangeable heme, suggesting formation of heme-inhibitor complexes in the parasite. In an effort to confirm this, the presence of a Br atom in one of the benzimidazole derivatives was exploited to image its distribution in the parasite using electron spectroscopic imaging of Br, an element not naturally abundant in cells. This showed that the compound colocalized with iron, consistent with its presence as a heme complex. Direct evidence for this complex was then obtained using confocal Raman microscopy. Exchangeable heme and inhibitor were found to increase with decreased rate of killing, suggesting that slow-acting compounds have more time to build up exchangeable heme complexes. Lastly, some but not all compounds evidently cause pro-oxidant effects because their activity could be attenuated with N-acetylcysteine and potentiated with t-butyl hydroperoxide. Collectively, these findings suggest that hemozoin inhibitors act as complexes with free heme, each with its own unique activity.


Subject(s)
Antimalarials , Hemeproteins , Antimalarials/pharmacology , Heme , Plasmodium falciparum
4.
Molecules ; 25(7)2020 Mar 29.
Article in English | MEDLINE | ID: mdl-32235391

ABSTRACT

With the continued loss of antimalarials to resistance, drug repositioning may have a role in maximising efficiency and accelerating the discovery of new antimalarial drugs. Bayesian statistics was previously used as a tool to virtually screen USFDA approved drugs for predicted ß-haematin (synthetic haemozoin) inhibition and in vitro antimalarial activity. Here, we report the experimental evaluation of nine of the highest ranked drugs, confirming the accuracy of the model by showing an overall 93% hit rate. Lapatinib, nilotinib, and lomitapide showed the best activity for inhibition of ß-haematin formation and parasite growth and were found to inhibit haemozoin formation in the parasite, providing mechanistic insights into their mode of antimalarial action. We then screened the USFDA approved drugs for binding to the ß-haematin crystal, applying a docking method in order to evaluate its performance. The docking method correctly identified imatinib, lapatinib, nilotinib, and lomitapide. Experimental evaluation of 22 of the highest ranked purchasable drugs showed a 24% hit rate. Lapatinib and nilotinib were chosen as templates for shape and electrostatic similarity screening for lead hopping using the in-stock ChemDiv compound catalogue. The actives were novel structures worthy of future investigation. This study presents a comparison of different in silico methods to identify new haemozoin-inhibiting chemotherapeutic alternatives for malaria that proved to be useful in different ways when taking into consideration their strengths and limitations.


Subject(s)
Antimalarials/pharmacology , Benzimidazoles/pharmacology , Hemeproteins/antagonists & inhibitors , Lapatinib/pharmacology , Plasmodium falciparum/drug effects , Pyrimidines/pharmacology , Antimalarials/chemistry , Benzimidazoles/chemistry , Binding Sites , Chloroquine/pharmacology , Drug Repositioning , Drug Resistance/drug effects , Erythrocytes/drug effects , Erythrocytes/parasitology , Hemeproteins/biosynthesis , Hemeproteins/chemistry , High-Throughput Screening Assays , Humans , Inhibitory Concentration 50 , Lapatinib/chemistry , Molecular Docking Simulation , Plasmodium falciparum/growth & development , Plasmodium falciparum/metabolism , Pyrimethamine/pharmacology , Pyrimidines/chemistry , Thermodynamics
5.
Sci Rep ; 10(1): 3374, 2020 02 25.
Article in English | MEDLINE | ID: mdl-32099045

ABSTRACT

Malaria remains a major public health problem. With the loss of antimalarials to resistance, the malaria burden will likely continue for decades. New antimalarial scaffolds are crucial to avoid cross-resistance. Here, we present the first structure based virtual screening using the ß-haematin crystal as a target for new inhibitor scaffolds by applying a docking method. The ZINC15 database was searched for compounds with high binding affinity with the surface of the ß-haematin crystal using the PyRx Virtual Screening Tool. Top-ranked compounds predicted to interact with ß-haematin were submitted to a second screen applying in silico toxicity and drug-likeness predictions using Osiris DataWarrior. Fifteen compounds were purchased for experimental testing. An NP-40 mediated ß-haematin inhibition assay and parasite growth inhibition activity assay were performed. The benzoxazole moiety was found to be a promising scaffold for further development, showing intraparasitic haemozoin inhibition using a cellular haem fractionation assay causing a decrease in haemozoin in a dose dependent manner with a corresponding increase in exchangeable haem. A ß-haematin inhibition hit rate of 73% was found, a large enrichment over random screening, demonstrating that virtual screening can be a useful and cost-effective approach in the search for new haemozoin inhibiting antimalarials.


Subject(s)
Antimalarials/pharmacology , Hemeproteins/antagonists & inhibitors , Plasmodium falciparum/drug effects , Protozoan Proteins/antagonists & inhibitors , Animals , Antimalarials/chemistry , Antimalarials/metabolism , Benzoxazoles/chemistry , Benzoxazoles/metabolism , Benzoxazoles/pharmacology , Binding Sites , CHO Cells , Cell Survival/drug effects , Cricetinae , Cricetulus , Databases, Protein , Hemeproteins/metabolism , Molecular Docking Simulation , Protein Conformation , Protozoan Proteins/metabolism
6.
Eur J Med Chem ; 159: 243-254, 2018 Nov 05.
Article in English | MEDLINE | ID: mdl-30296683

ABSTRACT

The 2-phenylbenzimidazole scaffold has recently been discovered to inhibit ß-hematin (synthetic hemozoin) formation by high throughput screening. Here, a library of 325,728 N-4-(1H-benzo[d]imidazol-2-yl)aryl)benzamides was enumerated, and Bayesian statistics used to predict ß-hematin and Plasmodium falciparum growth inhibition. Filtering predicted inactives and compounds with negligible aqueous solubility reduced the library to 35,124. Further narrowing to compounds with terminal aryl ring substituents only, reduced the library to 18, 83% of which were found to inhibit ß-hematin formation <100 µM and 50% parasite growth <2 µM. Four compounds showed nanomolar parasite growth inhibition activities, no cross-resistance in a chloroquine resistant strain and low cytotoxicity. QSAR analysis showed a strong association of parasite growth inhibition with inhibition of ß-hematin formation and the most active compound inhibited hemozoin formation in P. falciparum, with consequent increasing exchangeable heme. Pioneering use of molecular docking for this system demonstrated predictive ability and could rationalize observed structure activity trends.


Subject(s)
Antimalarials/pharmacology , Benzimidazoles/pharmacology , Hemeproteins/antagonists & inhibitors , Malaria, Falciparum/drug therapy , Plasmodium falciparum/drug effects , Antimalarials/chemical synthesis , Antimalarials/chemistry , Benzimidazoles/chemical synthesis , Benzimidazoles/chemistry , Dose-Response Relationship, Drug , Molecular Docking Simulation , Molecular Structure , Parasitic Sensitivity Tests , Structure-Activity Relationship
7.
Nat Microbiol ; 2(10): 1403-1414, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28808258

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.


Subject(s)
Antimalarials/pharmacology , Malaria, Falciparum/drug therapy , Malaria/drug therapy , Plasmodium berghei/drug effects , Quinolines/pharmacology , Amino Acid Sequence , Animals , Anopheles , CRISPR-Cas Systems/genetics , DNA, Protozoan/genetics , DNA, Protozoan/metabolism , Drug Combinations , Drug Resistance , Endocytosis/drug effects , Ethanolamines/pharmacology , Fluorenes/pharmacology , Gene Editing , HEK293 Cells , Heme , Hemoglobins/drug effects , High-Throughput Screening Assays , Humans , Lumefantrine , Malaria/transmission , Malaria, Falciparum/blood , Malaria, Falciparum/transmission , Male , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Multidrug Resistance-Associated Proteins/drug effects , Multidrug Resistance-Associated Proteins/genetics , Mutation , Oocysts/drug effects , Plasmodium berghei/pathogenicity , Plasmodium falciparum/drug effects , Plasmodium falciparum/genetics , Quinolines/chemistry
8.
mBio ; 8(3)2017 05 09.
Article in English | MEDLINE | ID: mdl-28487425

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.IMPORTANCE 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.


Subject(s)
Antimalarials/pharmacology , Drug Resistance/genetics , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics , Plasmodium falciparum/drug effects , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Quinolines/pharmacology , Antimalarials/chemistry , Artemisinins/therapeutic use , Cambodia , Gene Editing , Humans , Lethal Dose 50 , Malaria, Falciparum/epidemiology , Malaria, Falciparum/parasitology , Membrane Transport Proteins/metabolism , Multidrug Resistance-Associated Proteins/genetics , Mutation/drug effects , Plasmodium falciparum/genetics , Protein Isoforms , Protozoan Proteins/metabolism , Quinolines/chemistry
9.
ACS Med Chem Lett ; 8(2): 201-205, 2017 Feb 09.
Article in English | MEDLINE | ID: mdl-28197312

ABSTRACT

In a previous study, target based screening was carried out for inhibitors of ß-hematin (synthetic hemozoin) formation, and a series of triarylimidazoles were identified as active against Plasmodium falciparum. Here, we report the subsequent synthesis and testing of derivatives with varying substituents on the three phenyl rings for this series. The results indicated that a 2-hydroxy-1,3-dimethoxy substitution pattern on ring A is required for submicromolar parasite activity. In addition, cell-fractionation studies revealed uncommonly large, dose-dependent increases of P. falciparum intracellular exchangeable (free) heme, correlating with decreased parasite survival for ß-hematin inhibiting derivatives.

10.
PLoS Pathog ; 12(11): e1005976, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27832198

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.


Subject(s)
Drug Resistance/genetics , Genetic Fitness/genetics , Malaria, Falciparum/genetics , Membrane Transport Proteins/genetics , Plasmodium falciparum/physiology , Protozoan Proteins/genetics , Aminoquinolines/pharmacology , Antimalarials/pharmacology , Genotype , Humans , Mass Spectrometry , Microbial Sensitivity Tests , Mutation , Vacuoles/metabolism
11.
J Med Chem ; 59(13): 6512-30, 2016 07 14.
Article in English | MEDLINE | ID: mdl-27299916

ABSTRACT

Quinoline antimalarials target hemozoin formation causing a cytotoxic accumulation of ferriprotoporphyrin IX (Fe(III)PPIX). Well-developed SAR models exist for ß-hematin inhibition, parasite activity, and cellular mechanisms for this compound class, but no comparably detailed investigations exist for other hemozoin inhibiting chemotypes. Here, benzamide analogues based on previous HTS hits have been purchased or synthesized. Only derivatives containing an electron deficient aromatic ring and capable of adopting flat conformations, optimal for π-π interactions with Fe(III)PPIX, inhibited ß-hematin formation. The two most potent analogues showed nanomolar parasite activity, with little CQ cross-resistance, low cytotoxicity, and high in vitro microsomal stability. Selected analogues inhibited hemozoin formation in Plasmodium falciparum causing high levels of free heme. In contrast to quinolines, introduction of amine side chains did not lead to benzamide accumulation in the parasite. These data reveal complex relationships between heme binding, free heme levels, cellular accumulation, and in vitro activity of potential novel antimalarials.


Subject(s)
Antimalarials/pharmacology , Benzamides/pharmacology , Hemeproteins/antagonists & inhibitors , Plasmodium falciparum/drug effects , Antimalarials/chemical synthesis , Antimalarials/chemistry , Benzamides/chemical synthesis , Benzamides/chemistry , Dose-Response Relationship, Drug , Hemeproteins/metabolism , Molecular Structure , Parasitic Sensitivity Tests , Structure-Activity Relationship
12.
Malar J ; 14: 253, 2015 Jun 24.
Article in English | MEDLINE | ID: mdl-26099266

ABSTRACT

BACKGROUND: The activity of several well-known anti-malarials, including chloroquine (CQ), is attributed to their ability to inhibit the formation of haemozoin (Hz) in the malaria parasite. The formation of inert Hz, or malaria pigment, from toxic haem acquired from the host red blood cell of the parasite during haemoglobin digestion represents a pathway essential for parasite survival. Inhibition of this critical pathway therefore remains a desirable target for novel anti-malarials. A recent publication described the results of a haem fractionation assay used to directly determine haemoglobin, free haem and Hz in Plasmodium falciparum inoculated with CQ. CQ was shown to cause a dose-dependent increase in cellular-free haem that was correlated with decreased parasite survival. The method provided valuable information but was limited due to its low throughput and high demand on parasite starting material. Here, this haem fractionation assay has been successfully adapted to a higher throughput method in 24-well plates, significantly reducing lead times and starting material volumes. METHODS: All major haem species in P. falciparum trophozoites, isolated through a series of cellular fractionation steps were determined spectrophotometrically in aqueous pyridine (5 % v/v, pH 7.5) as a low spin complex with haematin. Cell counts were determined using a haemocytometer and a rapid novel fluorescent flow cytometry method. RESULTS: A higher throughput haem fractionation assay in 24-well plates, containing at most ten million trophozoites was validated against the original published method using CQ and its robustness was confirmed. It provided a minimum six-fold improvement in productivity and 24-fold reduction in starting material volume. The assay was successfully applied to amodiaquine (AQ), which was shown to inhibit Hz formation, while the antifolate pyrimethamine (PYR) and the mitochondrial electron transporter inhibitor atovaquone (Atov) demonstrated no increase in toxic cellular free haem. CONCLUSIONS: This higher throughput cellular haem fractionation assay can easily be applied to novel anti-malarials with a significantly decreased lead time, providing a valuable tool with which to probe the mechanisms of action of both new and established anti-malarials.


Subject(s)
Antimalarials/pharmacology , Colorimetry/methods , Heme/analysis , Plasmodium falciparum/drug effects , Trophozoites/drug effects , Amodiaquine/pharmacology , Atovaquone/pharmacology , Chloroquine/pharmacology , Pyrimethamine/pharmacology
13.
Bioorg Med Chem ; 23(16): 5210-7, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-25573118

ABSTRACT

A large quantity of high throughput screening (HTS) data for antimalarial activity has become available in recent years. This includes both phenotypic and target-based activity. Realising the maximum value of these data remains a challenge. In this respect, methods that allow such data to be used for virtual screening maximise efficiency and reduce costs. In this study both in vitro antimalarial activity and inhibitory data for ß-haematin formation, largely obtained from publically available sources, has been used to develop Bayesian models for inhibitors of ß-haematin formation and in vitro antimalarial activity. These models were used to screen two in silico compound libraries. In the first, the 1510 U.S. Food and Drug Administration approved drugs available on PubChem were ranked from highest to lowest Bayesian score based on a training set of ß-haematin inhibiting compounds active against Plasmodium falciparum that did not include any of the clinical antimalarials or close analogues. The six known clinical antimalarials that inhibit ß-haematin formation were ranked in the top 2.1% of compounds. Furthermore, the in vitro antimalarial hit-rate for this prioritised set of compounds was found to be 81% in the case of the subset where activity data are available in PubChem. In the second, a library of about 5000 commercially available compounds (Aldrich(CPR)) was virtually screened for ability to inhibit ß-haematin formation and then for in vitro antimalarial activity. A selection of 34 compounds was purchased and tested, of which 24 were predicted to be ß-haematin inhibitors. The hit rate for inhibition of ß-haematin formation was found to be 25% and a third of these were active against P. falciparum, corresponding to enrichments estimated at about 25- and 140-fold relative to random screening, respectively.


Subject(s)
Antimalarials/chemistry , Antimalarials/pharmacology , Drug Discovery/methods , Hemeproteins/antagonists & inhibitors , Machine Learning , Malaria, Falciparum/drug therapy , Plasmodium falciparum/drug effects , Bayes Theorem , Databases, Pharmaceutical , Hemeproteins/metabolism , Humans , Malaria, Falciparum/parasitology , Models, Biological , Parasitic Sensitivity Tests/methods , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology
14.
ACS Chem Biol ; 8(1): 133-7, 2013 Jan 18.
Article in English | MEDLINE | ID: mdl-23043646

ABSTRACT

By using cell fractionation and measurement of Fe(III)heme-pyridine, the antimalarial chloroquine (CQ) has been shown to cause a dose-dependent decrease in hemozoin and concomitant increase in toxic free heme in cultured Plasmodium falciparum that is directly correlated with parasite survival. Transmission electron microscopy techniques have further shown that heme is redistributed from the parasite digestive vacuole to the cytoplasm and that CQ disrupts hemozoin crystal growth, resulting in mosaic boundaries in the crystals formed in the parasite. Extension of the cell fractionation study to other drugs has shown that artesunate, amodiaquine, lumefantrine, mefloquine, and quinine, all clinically important antimalarials, also inhibit hemozoin formation in the parasite cell, while the antifolate pyrimethamine and its combination with sulfadoxine do not. This study finally provides direct evidence in support of the hemozoin inhibition hypothesis for the mechanism of action of CQ and shows that other quinoline and related antimalarials inhibit cellular hemozoin formation.


Subject(s)
Antimalarials/pharmacology , Heme/physiology , Plasmodium falciparum/drug effects , Trophozoites/drug effects , Chloroquine/pharmacology , Dose-Response Relationship, Drug , Microscopy, Electron, Transmission
15.
Biochem J ; 365(Pt 2): 343-7, 2002 Jul 15.
Article in English | MEDLINE | ID: mdl-12033986

ABSTRACT

Chemical analysis has shown that Plasmodium falciparum trophozoites contain 61+/-2% of the iron within parasitized erythrocytes, of which 92+/-6% is located within the food vacuole. Of this, 88+/-9% is in the form of haemozoin. (57)Fe-Mössbauer spectroscopy shows that haemozoin is the only detectable iron species in trophozoites. Electron spectroscopic imaging confirms this conclusion.


Subject(s)
Heme/metabolism , Iron/metabolism , Plasmodium falciparum/metabolism , Animals , Hemeproteins/metabolism , Spectroscopy, Mossbauer
SELECTION OF CITATIONS
SEARCH DETAIL
...