Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-31608711

ABSTRACT

Objective: To conduct a phase-II trial using a ranking and selection paradigm where multiple treatments are compared with limited sample size and the best is chosen for a subsequent efficacy trial versus placebo. This strategy can find an effective treatment faster than traditional strategy of conducting larger trials against placebo. Methods: Sixty amyotrophic lateral sclerosis (ALS) participants were randomized 1:1:1 to creatine 30 g/day (CRE), tamoxifen 40 mg/day (T40), or tamoxifen 80 mg/day (T80), with matching placebo. The primary outcome was 38-week change in ALS Functional Rating Scale-Revised (ALSFRS-R), analyzed in a repeated-measures ANOVA. Secondary outcomes included slow vital capacity (SVC), quantitative muscle strength, early drug discontinuation (EDD), adverse events (AEs), and survival. Results: CRE participants experienced higher rates of drug-related AEs (82% vs. 43% T40, 47% T80) and EDD (50% vs. 24% T40, 29% T80). T80 participants experienced slower adjusted mean decline in ALSFRS-R in points/month (-0.80 vs. -0.84 T40, -0.85 CRE) and quantitative muscle strength but not in SVC and higher rates of mortality. Conclusion: Efficacy of T80 ranked numerically superior to CRE and T40 with respect to ALSFRS-R decline. Following the selection paradigm, T80 would be chosen to test against placebo. The approach was not designed to distinguish among treatments that are nearly equally effective or ineffective. If treatments are equivalent, then under the paradigm, it does not matter which treatment is selected. Newer approaches for increasing trial efficiency, including an adaptive platform trial design, may mitigate limitations of the selection design.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Creatine/therapeutic use , Tamoxifen/administration & dosage , Tamoxifen/therapeutic use , Adult , Aged , Creatine/adverse effects , Double-Blind Method , Female , Humans , Male , Middle Aged , Muscle Strength , Vital Capacity/drug effects , Vital Capacity/physiology
2.
Nat Microbiol ; 2: 17031, 2017 Mar 13.
Article in English | MEDLINE | ID: mdl-28288098

ABSTRACT

Malaria control is heavily dependent on chemotherapeutic agents for disease prevention and drug treatment. Defining the mechanism of action for licensed drugs, for which no target is characterized, is critical to the development of their second-generation derivatives to improve drug potency towards inhibition of their molecular targets. Mefloquine is a widely used antimalarial without a known mode of action. Here, we demonstrate that mefloquine is a protein synthesis inhibitor. We solved a 3.2 Šcryo-electron microscopy structure of the Plasmodium falciparum 80S ribosome with the (+)-mefloquine enantiomer bound to the ribosome GTPase-associated centre. Mutagenesis of mefloquine-binding residues generates parasites with increased resistance, confirming the parasite-killing mechanism. Furthermore, structure-guided derivatives with an altered piperidine group, predicted to improve binding, show enhanced parasiticidal effect. These data reveal one possible mode of action for mefloquine and demonstrate the vast potential of cryo-electron microscopy to guide the development of mefloquine derivatives to inhibit parasite protein synthesis.


Subject(s)
Antimalarials/pharmacology , Mefloquine/pharmacology , Plasmodium falciparum/drug effects , Protein Biosynthesis/drug effects , Protein Synthesis Inhibitors/pharmacology , Ribosomes/drug effects
3.
Mol Microbiol ; 96(4): 796-814, 2015 May.
Article in English | MEDLINE | ID: mdl-25689481

ABSTRACT

The malaria parasite Plasmodium falciparum has two translationally active organelles - the apicoplast and mitochondrion, which import nuclear-encoded translation factors to mediate protein synthesis. Initiation of translation is a complex step wherein initiation factors (IFs) act in a regulated manner to form an initiation complex. We identified putative organellar IFs and investigated the targeting, structure and function of IF1, IF2 and IF3 homologues encoded by the parasite nuclear genome. A single PfIF1 is targeted to the apicoplast. Apart from its critical ribosomal interactions, PfIF1 also exhibited nucleic-acid binding and melting activities and mediated transcription anti-termination. This suggests a prominent ancillary function for PfIF1 in destabilisation of DNA and RNA hairpin loops encountered during transcription and translation of the A+T rich apicoplast genome. Of the three putative IF2 homologues, only one (PfIF2a) was an organellar protein with mitochondrial localisation. We additionally identified an IF3 (PfIF3a) that localised exclusively to the mitochondrion and another protein, PfIF3b, that was apicoplast targeted. PfIF3a exhibited ribosome anti-association activity, and monosome splitting by PfIF3a was enhanced by ribosome recycling factor (PfRRF2) and PfEF-G(Mit). These results fill a gap in our understanding of organellar translation in Plasmodium, which is the site of action of several anti-malarial compounds.


Subject(s)
Apicoplasts/genetics , Eukaryotic Initiation Factor-2/metabolism , Eukaryotic Initiation Factor-3/metabolism , Mitochondria/genetics , Peptide Chain Initiation, Translational , Plasmodium falciparum/genetics , Protozoan Proteins/metabolism , Apicoplasts/metabolism , Eukaryotic Initiation Factor-1/genetics , Eukaryotic Initiation Factor-1/metabolism , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-3/genetics , Mitochondria/metabolism , Plasmodium falciparum/metabolism , Protein Transport , Protozoan Proteins/genetics , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Sequence Alignment
4.
Int J Parasitol Drugs Drug Resist ; 4(1): 1-13, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24596663

ABSTRACT

Aminoacyl-tRNA synthetases are central enzymes in protein translation, providing the charged tRNAs needed for appropriate construction of peptide chains. These enzymes have long been pursued as drug targets in bacteria and fungi, but the past decade has seen considerable research on aminoacyl-tRNA synthetases in eukaryotic parasites. Existing inhibitors of bacterial tRNA synthetases have been adapted for parasite use, novel inhibitors have been developed against parasite enzymes, and tRNA synthetases have been identified as the targets for compounds in use or development as antiparasitic drugs. Crystal structures have now been solved for many parasite tRNA synthetases, and opportunities for selective inhibition are becoming apparent. For different biological reasons, tRNA synthetases appear to be promising drug targets against parasites as diverse as Plasmodium (causative agent of malaria), Brugia (causative agent of lymphatic filariasis), and Trypanosoma (causative agents of Chagas disease and human African trypanosomiasis). Here we review recent developments in drug discovery and target characterisation for parasite aminoacyl-tRNA synthetases.

5.
PLoS One ; 8(9): e74408, 2013.
Article in English | MEDLINE | ID: mdl-24058559

ABSTRACT

The two organelles, apicoplast and mitochondrion, of the malaria parasite Plasmodium falciparum have unique morphology in liver and blood stages; they undergo complex branching and looping prior to division and segregation into daughter merozoites. Little is known about the molecular processes and proteins involved in organelle biogenesis in the parasite. We report the identification of an AAA+/FtsH protease homolog (PfFtsH1) that exhibits ATP- and Zn(2+)-dependent protease activity. PfFtsH1 undergoes processing, forms oligomeric assemblies, and is associated with the membrane fraction of the parasite cell. Generation of a transfectant parasite line with hemagglutinin-tagged PfFtsH1, and immunofluorescence assay with anti-PfFtsH1 Ab demonstrated that the protein localises to P. falciparum mitochondria. Phylogenetic analysis and the single transmembrane region identifiable in PfFtsH1 suggest that it is an i-AAA like inner mitochondrial membrane protein. Expression of PfFtsH1 in Escherichia coli converted a fraction of bacterial cells into division-defective filamentous forms implying a sequestering effect of the Plasmodium factor on the bacterial homolog, indicative of functional conservation with EcFtsH. These results identify a membrane-associated mitochondrial AAA+/FtsH protease as a candidate regulatory protein for organelle biogenesis in P. falciparum.


Subject(s)
Mitochondria/enzymology , Peptide Hydrolases/metabolism , Plasmodium falciparum/enzymology , Protozoan Proteins/metabolism , Adenosine Triphosphate/pharmacology , Animals , Cytokinesis/drug effects , Escherichia coli/metabolism , Fluorescent Antibody Technique , Membrane Proteins/metabolism , Mitochondria/drug effects , Parasites/cytology , Parasites/drug effects , Parasites/enzymology , Peptide Hydrolases/chemistry , Phylogeny , Plasmodium falciparum/cytology , Plasmodium falciparum/drug effects , Protein Processing, Post-Translational/drug effects , Protein Structure, Quaternary , Protein Transport/drug effects , Protozoan Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Zinc/pharmacology
6.
Mol Microbiol ; 88(5): 891-905, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23614815

ABSTRACT

The reduced genomes of the apicoplast and mitochondrion of the malaria parasite Plasmodium falciparum are actively translated and antibiotic-mediated translation inhibition is detrimental to parasite survival. In order to understand recycling of organellar ribosomes, a critical step in protein translation, we identified ribosome recycling factors (RRF) encoded by the parasite nuclear genome. Targeting of PfRRF1 and PfRRF2 to the apicoplast and mitochondrion respectively was established by localization of leader sequence-GFP fusions. Unlike any RRF characterized thus far, PfRRF2 formed dimers with disulphide interaction(s) and additionally localized in the cytoplasm, thus suggesting adjunct functions for the factor. PfRRF1 carries a large 108-amino-acid insertion in the functionally critical hinge region between the head and tail domains of the protein, yet complemented Escherichia coli RRF in the LJ14frr(ts) mutant and disassembled surrogate E. coli 70S ribosomes in the presence of apicoplast-targeted EF-G. Recombinant PfRRF2 bound E. coli ribosomes and could split monosomes in the presence of the relevant mitochondrial EF-G but failed to complement the LJ14frr(ts) mutant. Although proteins comprising subunits of P. falciparum organellar ribosomes are predicted to differ from bacterial and mitoribosomal counterparts, our results indicate that the essential interactions required for recycling are conserved in parasite organelles.


Subject(s)
Apicoplasts/enzymology , Apicoplasts/genetics , Mitochondria/enzymology , Mitochondria/genetics , Plasmodium falciparum/enzymology , Plasmodium falciparum/genetics , Ribosomal Proteins/genetics , Amino Acid Sequence , Molecular Sequence Data , Protein Biosynthesis , Protein Multimerization , Protein Transport , RNA, Messenger/metabolism , Ribosomal Proteins/chemistry , Ribosomal Proteins/metabolism , Ribosomes/metabolism , Sequence Alignment
7.
Int J Parasitol ; 42(2): 177-86, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22222968

ABSTRACT

The causative agent of malaria, Plasmodium, possesses three translationally active compartments: the cytosol, the mitochondrion and a relic plastid called the apicoplast. Aminoacyl-tRNA synthetases to charge tRNA are thus required for all three compartments. However, the Plasmodiumfalciparum genome encodes too few tRNA synthetases to supply a unique enzyme for each amino acid in all three compartments. We have investigated the subcellular localisation of three tRNA synthetases (AlaRS, GlyRS and ThrRS), which occur only once in the nuclear genome, and we show that each of these enzymes is dually localised to the P. falciparum cytosol and the apicoplast. No mitochondrial fraction is apparent for these three enzymes, which suggests that the Plasmodium mitochondrion lacks at least these three tRNA synthetases. The unique Plasmodium ThrRS is the presumed target of the antimalarial compound borrelidin. Borrelidin kills P. falciparum parasites quickly without the delayed death effect typical of apicoplast translation inhibitors and without an observable effect on apicoplast morphology. By contrast, mupirocin, an inhibitor of the apicoplast IleRS, kills with a delayed death effect that inhibits apicoplast growth and division. Because inhibition of dual targeted tRNA synthetases should arrest translation in all compartments of the parasite, these enzymes deserve further investigation as potential targets for antimalarial drug development.


Subject(s)
Amino Acyl-tRNA Synthetases/metabolism , Cytosol/enzymology , Plasmodium falciparum/physiology , Plastids/enzymology , Antimalarials/pharmacology , Fatty Alcohols/pharmacology , Mupirocin/pharmacology , Plasmodium falciparum/metabolism , Protein Transport
8.
Trends Parasitol ; 27(10): 467-76, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21741312

ABSTRACT

The protein translation machinery of the parasite Plasmodium is the target of important anti-malarial drugs, and encompasses many promising targets for future drugs. Plasmodium parasites have three subcellular compartments that house genomes; the nucleus, mitochondrion and apicoplast, and each requires its own compartmentalized transcription and translation apparatus for survival. Despite the availability of the complete genome sequence that should reveal the requisite elements for all three compartments, our understanding of the translation machineries is patchy. We review what is known about cytosolic and organellar translation in Plasmodium and discuss the molecules that have been identified through genome sequencing and post-genomic analysis. Some translation components are yet to be found in Plasmodium, whereas others appear to be shared between translationally active organelles.


Subject(s)
Genes, Protozoan , Plasmodium/metabolism , Protein Biosynthesis , Amino Acyl-tRNA Synthetases/metabolism , Antimalarials/pharmacology , Cell Nucleus/genetics , Cell Nucleus/metabolism , Gene Expression Regulation , Genome, Mitochondrial , Humans , Malaria/drug therapy , Malaria/parasitology , Mitochondria/drug effects , Mitochondria/genetics , Mitochondria/metabolism , Plasmodium/drug effects , Plasmodium/genetics , Plasmodium/growth & development , Protein Processing, Post-Translational , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , RNA, Transfer/genetics , RNA, Transfer/metabolism , Ribosomes/genetics , Ribosomes/metabolism
9.
Circ Res ; 109(4): 365-73, 2011 Aug 05.
Article in English | MEDLINE | ID: mdl-21700932

ABSTRACT

RATIONALE: Antibody-targeted delivery of imaging agents can enhance the sensitivity and accuracy of current imaging techniques. Similarly, homing of effector cells to disease sites increases the efficacy of regenerative cell therapy while reducing the number of cells required. Currently, targeting can be achieved via chemical conjugation to specific antibodies, which typically results in the loss of antibody functionality and in severe cell damage. An ideal conjugation technique should ensure retention of antigen-binding activity and functionality of the targeted biological component. OBJECTIVE: To develop a biochemically robust, highly reproducible, and site-specific coupling method using the Staphylococcus aureus sortase A enzyme for the conjugation of a single-chain antibody (scFv) to nanoparticles and cells for molecular imaging and cell homing in cardiovascular diseases. This scFv specifically binds to activated platelets, which play a pivotal role in thrombosis, atherosclerosis, and inflammation. METHODS AND RESULTS: The conjugation procedure involves chemical and enzyme-mediated coupling steps. The scFv was successfully conjugated to iron oxide particles (contrast agents for magnetic resonance imaging) and to model cells. Conjugation efficiency ranged between 50% and 70%, and bioactivity of the scFv after coupling was preserved. The targeting of scFv-coupled cells and nanoparticles to activated platelets was strong and specific as demonstrated in in vitro static adhesion assays, in a flow chamber system, in mouse intravital microscopy, and in in vivo magnetic resonance imaging of mouse carotid arteries. CONCLUSIONS: This unique biotechnological approach provides a versatile and broadly applicable tool for procuring targeted regenerative cell therapy and targeted molecular imaging in cardiovascular and inflammatory diseases and beyond.


Subject(s)
Aminoacyltransferases/metabolism , Bacterial Proteins/metabolism , Cell Movement , Cell Tracking/methods , Contrast Media , Cysteine Endopeptidases/metabolism , Magnetic Resonance Imaging , Magnetite Nanoparticles , Molecular Probe Techniques , Single-Chain Antibodies/metabolism , Thrombosis/pathology , Aminoacyltransferases/biosynthesis , Aminoacyltransferases/genetics , Animals , Bacterial Proteins/biosynthesis , Bacterial Proteins/genetics , Blood Platelets/metabolism , CHO Cells , Chlorides , Cricetinae , Cricetulus , Cysteine Endopeptidases/biosynthesis , Cysteine Endopeptidases/genetics , Disease Models, Animal , Ferric Compounds , Flow Cytometry , Green Fluorescent Proteins/metabolism , Humans , Mice , Mice, Inbred C57BL , Microscopy, Video , Platelet Activation , Recombinant Fusion Proteins/metabolism , Single-Chain Antibodies/biosynthesis , Single-Chain Antibodies/genetics , Thrombosis/chemically induced , Thrombosis/metabolism
10.
Lancet Neurol ; 9(5): 481-8, 2010 May.
Article in English | MEDLINE | ID: mdl-20363190

ABSTRACT

BACKGROUND: In a pilot study, lithium treatment slowed progression of amyotrophic lateral sclerosis (ALS). We aimed to confirm or disprove these findings by assessing the safety and efficacy of lithium in combination with riluzole in patients with ALS. METHODS: We did a double-blind, placebo-controlled trial with a time-to-event design. Between January and June, 2009, patients with ALS who were taking a stable dose of riluzole for at least 30 days were randomly assigned (1:1) by a centralised computer to receive either lithium or placebo. Patients, caregivers, investigators, and all site study staff with the exception of site pharmacists were masked to treatment assignment. The primary endpoint was the time to an event, defined as a decrease of at least six points on the revised ALS functional rating scale score or death. Interim analyses were planned for when 84 patients had been allocated treatment, 6 months later or after 55 events, and after 100 events. Analysis was by intention to treat. The stopping boundary for futility at the first interim analysis was a p value of at least 0.68. We used a log-rank test to compare the distributions of the time to an event between the lithium and placebo groups. This trial is registered with ClinicalTrials.gov, NCT00818389. FINDINGS: At the first interim analysis, 22 of 40 patients in the lithium group had an event compared with 20 of 44 patients in the placebo group (log rank p=0.51). The hazard ratio of reaching the primary endpoint was 1.13 (95% CI 0.61-2.07). The study was stopped at the first interim analysis because criterion for futility was met (p=0.78). The difference in mean decline in the ALS functional rating scale score between the lithium group and the placebo group was 0.15 (95% CI -0.43 to 0.73, p=0.61). There were no major safety concerns. Falls (p=0.04) and back pain (p=0.05) were more common in the lithium group than in the placebo group. INTERPRETATION: We found no evidence that lithium in combination with riluzole slows progression of ALS more than riluzole alone. The time-to-event endpoint and use of prespecified interim analyses enabled a clear result to be obtained rapidly. This design should be considered for future trials testing the therapeutic efficacy of drugs that are easily accessible to people with ALS. FUNDING: National Institute of Neurological Disorders and Stroke, ALS Association, and ALS Society of Canada.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Lithium/adverse effects , Medical Futility , Riluzole/adverse effects , Adult , Aged , Disease Progression , Double-Blind Method , Drug Therapy, Combination , Drug-Related Side Effects and Adverse Reactions , Humans , Intention to Treat Analysis , Lithium/administration & dosage , Male , Middle Aged , Patient Selection , Prognosis , Riluzole/administration & dosage , Treatment Outcome
11.
Clin Infect Dis ; 50(5): 779-86, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-20121574

ABSTRACT

BACKGROUND: We sought to use data captured in the electronic health record (EHR) to develop and validate a prediction rule for virologic failure among patients being treated for infection with human immunodeficiency virus (HIV). METHODS: We used EHRs at 2 Boston tertiary care hospitals, Massachusetts General Hospital and Brigham and Women's Hospital, to identify HIV-infected patients who were virologically suppressed (HIV RNA level < or = 400 copies/mL) on antiretroviral therapy (ART) during the period from 1 January 2005 through 31 December 2006. We used a multivariable logistic model with data from Massachusetts General Hospital to derive a 1-year virologic failure prediction rule. The model was validated using data from Brigham and Women's Hospital.We then simplified the scoring scheme to develop a clinical prediction rule. RESULTS: The 1-year virologic failure prediction model, using data from 712 patients from Massachusetts General Hospital, demonstrated good discrimination (C statistic, 0.78) and calibration (chi(2)= 6.6; P= .58). The validation model, based on 362 patients from Brigham and Women's Hospital, also showed good discrimination (C statistic, 0.79) and calibration (chi(2)= 1.9; P= .93). The clinical prediction rule included 7 predictors (suboptimal adherence, CD4 cell count < 100 cells/microL, drug and/or alcohol abuse, highly ART experienced, missed > or = 1 appointment, prior virologic failure, and suppressed < or = 12 months) and appropriately stratified patients in the validation data set into low-, medium-, and high-risk groups, with 1-year virologic failure rates of 3.0%, 13.0%, and 28.6%, respectively. CONCLUSIONS: A risk score based on 7 variables available in the EHR predicts HIV virologic failure at 1 year and could be used for targeted interventions to improve outcomes in HIV infection.


Subject(s)
Anti-HIV Agents/therapeutic use , Antiretroviral Therapy, Highly Active , HIV Infections/drug therapy , HIV Infections/virology , Viral Load , Adult , Boston , Data Collection/methods , Female , Humans , Male , Massachusetts , Medical Records Systems, Computerized , Middle Aged , Prognosis , Risk Factors , Treatment Failure
12.
Biochem J ; 403(1): 167-75, 2007 Apr 01.
Article in English | MEDLINE | ID: mdl-17155936

ABSTRACT

Plasmodium falciparum develops within the mature RBCs (red blood cells) of its human host in a PV (parasitophorous vacuole) that separates the host cell cytoplasm from the parasite surface. The pore-forming toxin, SLO (streptolysin O), binds to cholesterol-containing membranes and can be used to selectively permeabilize the host cell membrane while leaving the PV membrane intact. We found that in mixtures of infected and uninfected RBCs, SLO preferentially lyses uninfected RBCs rather than infected RBCs, presumably because of differences in cholesterol content of the limiting membrane. This provides a means of generating pure preparations of viable ring stage infected RBCs. As an alternative permeabilizing agent we have characterized EqtII (equinatoxin II), a eukaryotic pore-forming toxin that binds preferentially to sphingomyelin-containing membranes. EqtII lyses the limiting membrane of infected and uninfected RBCs with similar efficiency but does not disrupt the PV membrane. It generates pores of up to 100 nm, which allow entry of antibodies for immunofluorescence and immunogold labelling. The present study provides novel tools for the analysis of this important human pathogen and highlights differences between Plasmodium-infected and uninfected RBCs.


Subject(s)
Cell Membrane Permeability/drug effects , Cnidarian Venoms/pharmacology , Erythrocyte Membrane/physiology , Plasmodium falciparum/pathogenicity , Streptolysins/pharmacology , Animals , Bacterial Proteins/pharmacology , Cytotoxins/pharmacology , Erythrocyte Membrane/drug effects , Humans , Parasitemia/blood
13.
Mol Microbiol ; 54(1): 109-22, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15458409

ABSTRACT

The malaria parasite Plasmodium falciparum induces a sixfold increase in the phospholipid content of infected erythrocytes during its intraerythrocytic growth. We have characterized the lipid environments in parasitized erythrocyte using the hydrophobic probe, Nile Red. Spectral imaging with a confocal microscope revealed heterogeneous lipid environments in parasite-infected erythrocytes. An insight into the nature of these environments was gained by comparing these spectra with those of triacylglycerol/phospholipid emulsions and phospholipid membranes. Using this approach, we identified a population of intensely stained particles of a few hundred nanometers in size that are closely associated with the digestive vacuole of the parasite and appear to be composed of neutral lipids. Electron microscopy and isolation of food vacuoles confirmed the size of these particles and their intimate association respectively. Lipid analysis suggests that these neutral lipid bodies are composed of di- and triacylgycerols and may represent storage organelles for lipid intermediates that are generated during digestion of phospholipids in the food vacuole. Mono-, di- and triacylglycerol suspensions promote beta-haematin formation, suggesting that these neutral lipid bodies, or their precursors, may also be involved in haem detoxification. We also characterized other compartments of the infected erythrocyte that were stained less intensely with the Nile Red probe. Both the erythrocyte membrane and the parasite membrane network exhibit red shifts compared with the neutral lipid bodies that are consistent with cholesterol-rich and cholesterol-poor membranes respectively. Ratiometric imaging revealed more subtle variations in the lipid environments within the parasite membrane network.


Subject(s)
Diglycerides/analysis , Erythrocytes/parasitology , Plasmodium falciparum/metabolism , Triglycerides/analysis , Vacuoles/chemistry , Animals , Erythrocytes/ultrastructure , Malaria, Falciparum/parasitology , Microscopy, Confocal , Microscopy, Fluorescence , Oxazines , Plasmodium falciparum/growth & development , Vacuoles/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...