Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Int J Dermatol ; 62(9): e484-e486, 2023 09.
Article in English | MEDLINE | ID: mdl-36841896

Subject(s)
Dermatologists , Humans , Mutation
2.
6.
Sci Rep ; 5: 15930, 2015 Nov 06.
Article in English | MEDLINE | ID: mdl-26541648

ABSTRACT

The spread of Plasmodium falciparum multidrug resistance highlights the urgency to discover new targets and chemical scaffolds. Unfortunately, lack of experimentally validated functional information about most P. falciparum genes remains a strategic hurdle. Chemogenomic profiling is an established tool for classification of drugs with similar mechanisms of action by comparing drug fitness profiles in a collection of mutants. Inferences of drug mechanisms of action and targets can be obtained by associations between shifts in drug fitness and specific genetic changes in the mutants. In this screen, P. falciparum, piggyBac single insertion mutants were profiled for altered responses to antimalarial drugs and metabolic inhibitors to create chemogenomic profiles. Drugs targeting the same pathway shared similar response profiles and multiple pairwise correlations of the chemogenomic profiles revealed novel insights into drugs' mechanisms of action. A mutant of the artemisinin resistance candidate gene - "K13-propeller" gene (PF3D7_1343700) exhibited increased susceptibility to artemisinin drugs and identified a cluster of 7 mutants based on similar enhanced responses to the drugs tested. Our approach of chemogenomic profiling reveals artemisinin functional activity, linked by the unexpected drug-gene relationships of these mutants, to signal transduction and cell cycle regulation pathways.


Subject(s)
Antimalarials/pharmacology , Plasmodium falciparum/drug effects , Drug Discovery/methods , Drug Resistance, Multiple/drug effects , Drug Resistance, Multiple/genetics , Mutagenesis, Insertional/drug effects , Plasmodium falciparum/genetics , Protozoan Proteins/genetics
7.
Eukaryot Cell ; 12(9): 1171-8, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23813392

ABSTRACT

Intraerythrocytic development of the human malaria parasite Plasmodium falciparum appears as a continuous flow through growth and proliferation. To develop a greater understanding of the critical regulatory events, we utilized piggyBac insertional mutagenesis to randomly disrupt genes. Screening a collection of piggyBac mutants for slow growth, we isolated the attenuated parasite C9, which carried a single insertion disrupting the open reading frame (ORF) of PF3D7_1305500. This gene encodes a protein structurally similar to a mitogen-activated protein kinase (MAPK) phosphatase, except for two notable characteristics that alter the signature motif of the dual-specificity phosphatase domain, suggesting that it may be a low-activity phosphatase or pseudophosphatase. C9 parasites demonstrated a significantly lower growth rate with delayed entry into the S/M phase of the cell cycle, which follows the stage of maximum PF3D7_1305500 expression in intact parasites. Genetic complementation with the full-length PF3D7_1305500 rescued the wild-type phenotype of C9, validating the importance of the putative protein phosphatase PF3D7_1305500 as a regulator of pre-S-phase cell cycle progression in P. falciparum.


Subject(s)
Merozoites/growth & development , Mitogen-Activated Protein Kinase Phosphatases/metabolism , Mitosis , Plasmodium falciparum/enzymology , Protozoan Proteins/metabolism , S Phase , Amino Acid Motifs , Amino Acid Sequence , Catalytic Domain , Ecthyma, Contagious , Genes, Protozoan , Merozoites/enzymology , Mitogen-Activated Protein Kinase Phosphatases/chemistry , Mitogen-Activated Protein Kinase Phosphatases/genetics , Molecular Sequence Data , Mutagenesis, Insertional , Plasmodium falciparum/genetics , Plasmodium falciparum/growth & development , Protozoan Proteins/chemistry , Protozoan Proteins/genetics
8.
PLoS Genet ; 9(2): e1003305, 2013.
Article in English | MEDLINE | ID: mdl-23437009

ABSTRACT

In the G1 phase of the cell division cycle, eukaryotic cells prepare many of the resources necessary for a new round of growth including renewal of the transcriptional and protein synthetic capacities and building the machinery for chromosome replication. The function of G1 has an early evolutionary origin and is preserved in single and multicellular organisms, although the regulatory mechanisms conducting G1 specific functions are only understood in a few model eukaryotes. Here we describe a new G1 mutant from an ancient family of apicomplexan protozoans. Toxoplasma gondii temperature-sensitive mutant 12-109C6 conditionally arrests in the G1 phase due to a single point mutation in a novel protein containing a single RNA-recognition-motif (TgRRM1). The resulting tyrosine to asparagine amino acid change in TgRRM1 causes severe temperature instability that generates an effective null phenotype for this protein when the mutant is shifted to the restrictive temperature. Orthologs of TgRRM1 are widely conserved in diverse eukaryote lineages, and the human counterpart (RBM42) can functionally replace the missing Toxoplasma factor. Transcriptome studies demonstrate that gene expression is downregulated in the mutant at the restrictive temperature due to a severe defect in splicing that affects both cell cycle and constitutively expressed mRNAs. The interaction of TgRRM1 with factors of the tri-SNP complex (U4/U6 & U5 snRNPs) indicate this factor may be required to assemble an active spliceosome. Thus, the TgRRM1 family of proteins is an unrecognized and evolutionarily conserved class of splicing regulators. This study demonstrates investigations into diverse unicellular eukaryotes, like the Apicomplexa, have the potential to yield new insights into important mechanisms conserved across modern eukaryotic kingdoms.


Subject(s)
Alternative Splicing/genetics , Cell Cycle/genetics , RNA, Messenger , RNA-Binding Proteins , Toxoplasma , Conserved Sequence/genetics , G1 Phase/genetics , Gene Expression Regulation , Humans , Mutation , Nucleotide Motifs/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Sequence Homology, Amino Acid , Temperature , Toxoplasma/genetics , Toxoplasma/metabolism
9.
PLoS One ; 7(7): e40416, 2012.
Article in English | MEDLINE | ID: mdl-22792308

ABSTRACT

Mechanisms of drug resistance in Plasmodium vivax have been difficult to study partially because of the difficulties in culturing the parasite in vitro. This hampers monitoring drug resistance and research to develop or evaluate new drugs. There is an urgent need for a novel method to study mechanisms of P. vivax drug resistance. In this paper we report the development and application of the first Plasmodium falciparum expression system to stably express P. vivax dhfr-ts alleles. We used the piggyBac transposition system for the rapid integration of wild-type, single mutant (117N) and quadruple mutant (57L/58R/61M/117T) pvdhfr-ts alleles into the P. falciparum genome. The majority (81%) of the integrations occurred in non-coding regions of the genome; however, the levels of pvdhfr transcription driven by the P. falciparum dhfr promoter were not different between integrants of non-coding and coding regions. The integrated quadruple pvdhfr mutant allele was much less susceptible to antifolates than the wild-type and single mutant pvdhfr alleles. The resistance phenotype was stable without drug pressure. All the integrated clones were susceptible to the novel antifolate JPC-2067. Therefore, the piggyBac expression system provides a novel and important tool to investigate drug resistance mechanisms and gene functions in P. vivax.


Subject(s)
Plasmodium falciparum/genetics , Plasmodium vivax/enzymology , Protozoan Proteins/genetics , Tetrahydrofolate Dehydrogenase/genetics , Thymidylate Synthase/genetics , Amino Acid Substitution , Antimalarials/pharmacology , Cells, Cultured , Drug Resistance , Erythrocytes/parasitology , Folic Acid Antagonists/pharmacology , Gene Dosage , Humans , Inhibitory Concentration 50 , Mutagenesis, Insertional , Plasmodium falciparum/drug effects , Plasmodium vivax/genetics , Proguanil/pharmacology , Protozoan Proteins/biosynthesis , Pyrimethamine/pharmacology , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Tetrahydrofolate Dehydrogenase/biosynthesis , Thymidylate Synthase/biosynthesis , Transfection , Triazines/pharmacology
10.
J Trop Med ; 2012: 829210, 2012.
Article in English | MEDLINE | ID: mdl-22649460

ABSTRACT

The genome sequence of the human malaria parasite, Plasmodium falciparum, was released almost a decade ago. A majority of the Plasmodium genome, however, remains annotated to code for hypothetical proteins with unknown functions. The introduction of forward genetics has provided novel means to gain a better understanding of gene functions and their associated phenotypes in Plasmodium. Even with certain limitations, the technique has already shown significant promise to increase our understanding of parasite biology needed for rationalized drug and vaccine design. Further improvements to the mutagenesis technique and the design of novel genetic screens should lead us to some exciting discoveries about the critical weaknesses of Plasmodium, and greatly aid in the development of new disease intervention strategies.

11.
Eukaryot Cell ; 10(9): 1257-63, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21803864

ABSTRACT

Coordinated regulation of gene expression is a hallmark of the Plasmodium falciparum asexual blood-stage development cycle. We report that carbon catabolite repressor protein 4 (CCR4)-associated factor 1 (CAF1) is critical in regulating more than 1,000 genes during malaria parasites' intraerythrocytic stages, especially egress and invasion proteins. CAF1 knockout results in mistimed expression, aberrant accumulation and localization of proteins involved in parasite egress, and invasion of new host cells, leading to premature release of predominantly half-finished merozoites, drastically reducing the intraerythrocytic growth rate of the parasite. This study demonstrates that CAF1 of the CCR4-Not complex is a significant gene regulatory mechanism needed for Plasmodium development within the human host.


Subject(s)
Erythrocytes/parasitology , Gene Deletion , Gene Expression , Host-Parasite Interactions/genetics , Plasmodium falciparum/genetics , Plasmodium falciparum/pathogenicity , Transcription Factors/genetics , Animals , Cell Proliferation , Erythrocytes/pathology , Gene Expression Regulation , Gene Knockout Techniques , Humans , Life Cycle Stages , Malaria, Falciparum/parasitology , Merozoites/metabolism , Oligonucleotide Array Sequence Analysis/methods , Plasmodium falciparum/growth & development , Transcription Factors/metabolism
12.
PLoS One ; 5(10): e13282, 2010 Oct 11.
Article in English | MEDLINE | ID: mdl-20949012

ABSTRACT

A majority of the Plasmodium falciparum genome codes for genes with unknown functions, which presents a major challenge to understanding the parasite's biology. Large-scale functional analysis of the parasite genome is essential to pave the way for novel therapeutic intervention strategies against the disease and yet difficulties in genetic manipulation of this deadly human malaria parasite have been a major hindrance for functional analysis of its genome. Here, we used a forward functional genomic approach to study P. falciparum and identify genes important for optimal parasite development in the disease-causing, intraerythrocytic stages. We analyzed 123 piggyBac insertion mutants of P. falciparum for proliferation efficiency in the intraerythrocytic stages, in vitro. Almost 50% of the analyzed mutants showed significant reduction in proliferation efficiency, with 20% displaying severe defects. Functional categorization of genes in the severely attenuated mutants revealed significant enrichment for RNA binding proteins, suggesting the significance of post-transcriptional gene regulation in parasite development and emphasizing its importance as an antimalarial target. This study demonstrates the feasibility of much needed forward genetics approaches for P. falciparum to better characterize its genome and accelerate drug and vaccine development.


Subject(s)
Erythrocytes/parasitology , Genes, Protozoan , Plasmodium falciparum/growth & development , Animals , Mutation , Plasmodium falciparum/genetics
13.
BMC Microbiol ; 9: 83, 2009 May 07.
Article in English | MEDLINE | ID: mdl-19422698

ABSTRACT

BACKGROUND: Much of the Plasmodium falciparum genome encodes hypothetical proteins with limited homology to other organisms. A lack of robust tools for genetic manipulation of the parasite limits functional analysis of these hypothetical proteins and other aspects of the Plasmodium genome. Transposon mutagenesis has been used widely to identify gene functions in many organisms and would be extremely valuable for functional analysis of the Plasmodium genome. RESULTS: In this study, we investigated the lepidopteran transposon, piggyBac, as a molecular genetic tool for functional characterization of the Plasmodium falciparum genome. Through multiple transfections, we generated 177 unique P. falciparum mutant clones with mostly single piggyBac insertions in their genomes. Analysis of piggyBac insertion sites revealed random insertions into the P. falciparum genome, in regards to gene expression in parasite life cycle stages and functional categories. We further explored the possibility of forward genetic studies in P. falciparum with a phenotypic screen for attenuated growth, which identified several parasite genes and pathways critical for intra-erythrocytic development. CONCLUSION: Our results clearly demonstrate that piggyBac is a novel, indispensable tool for forward functional genomics in P. falciparum that will help better understand parasite biology and accelerate drug and vaccine development.


Subject(s)
DNA Transposable Elements , Genome, Protozoan , Genomics/methods , Plasmodium falciparum/genetics , Animals , Mutagenesis, Insertional , Plasmids , Plasmodium falciparum/growth & development , Transfection
14.
Exp Parasitol ; 121(1): 110-4, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18950624

ABSTRACT

Strict regulation of gene expression is critical for the development of the malaria parasite within multiple host cell types. However, much remains unexplored regarding gene regulation in Plasmodium falciparum with only a few components of the gene regulation machinery identified thus far. Better characterization of transcript structures with precise mapping of transcript ends will greatly aid in the search of conserved regulatory sequences in the genome. Transcript analysis of maebl, a member of the ebl gene family, in P. falciparum intra-erythrocytic stages has revealed a unique transcript structure for maebl. The 5'-untranslated region of maebl transcript is exceptionally long (>2 kb) with a small multi-exon open reading frame, annotated as a putative mitochondrial ATP synthase (PF11_0485) in the Plasmodium database. Northern blot hybridizations and RT-PCR analysis confirmed a bicistronic message for maebl along with PF11_0485. We further identified the minimal maebl promoter to be upstream of PF11_0485 by using transient chloramphenicol acetyl transferase (CAT) reporter assays. The occurrence of a bicistronic mRNA in Plasmodium is both novel and unusual for a lower eukaryote and adds on to the complexity of gene regulation in malaria parasites.


Subject(s)
Plasmodium falciparum/genetics , Protozoan Proteins/chemistry , Receptors, Cell Surface/chemistry , Transcription, Genetic , Animals , Blotting, Northern , Cloning, Molecular , DNA Primers/chemistry , DNA, Protozoan/chemistry , DNA, Protozoan/isolation & purification , Erythrocytes/parasitology , Humans , Plasmodium falciparum/chemistry , Polymerase Chain Reaction , Promoter Regions, Genetic , Protozoan Proteins/genetics , RNA, Protozoan/chemistry , RNA, Protozoan/isolation & purification , Random Amplified Polymorphic DNA Technique , Receptors, Cell Surface/genetics
15.
Malar J ; 7: 222, 2008 Oct 29.
Article in English | MEDLINE | ID: mdl-18959784

ABSTRACT

BACKGROUND: Cloning of parasites by limiting dilution is an essential and rate-limiting step in many aspects of malaria research including genomic and genetic manipulation studies. The standard Giemsa-stained blood smears to detect parasites is time-consuming, whereas the more sensitive parasite lactate dehydrogenase assay involves multiple steps and requires fresh reagents. A simple PCR-based method was therefore tested for parasite detection that can be adapted to high throughput studies. METHODS: Approximately 1 microL of packed erythrocytes from each well of a microtiter cloning plate was directly used as template DNA for a PCR reaction with primers for the parasite 18s rRNA gene. Positive wells containing parasites were identified after rapid separation of PCR products by gel electrophoresis. RESULTS: The PCR-based method can consistently detect a parasitaemia as low as 0.0005%, which is equivalent to 30 parasite genomes in a single well of a 96-well plate. Parasite clones were easily detected from cloning plates using this method and a comparison of PCR results with Giemsa-stained blood smears showed that PCR not only detected all the positive wells identified in smears, but also detected wells not identified otherwise, thereby confirming its sensitivity. CONCLUSION: The PCR-based method reported here is a simple, sensitive and efficient method for detecting parasite clones in culture. This method requires very little manual labor and can be completely automated for high throughput studies. The method is sensitive enough to detect parasites a week before they can be seen in Giemsa smears and is highly effective in identifying slow growing parasite clones.


Subject(s)
Malaria, Falciparum/diagnosis , Plasmodium falciparum/genetics , Polymerase Chain Reaction/methods , Animals , Azure Stains , Clone Cells , Plasmodium falciparum/isolation & purification , RNA, Ribosomal, 18S/genetics , Sensitivity and Specificity
16.
PLoS Pathog ; 4(8): e1000118, 2008 Aug 08.
Article in English | MEDLINE | ID: mdl-18688278

ABSTRACT

Plasmodium falciparum is the protozoan parasite that causes the most virulent of human malarias. The blood stage parasites export several hundred proteins into their host erythrocyte that underlie modifications linked to major pathologies of the disease and parasite survival in the blood. Unfortunately, most are 'hypothetical' proteins of unknown function, and those that are essential for parasitization of the erythrocyte cannot be 'knocked out'. Here, we combined bioinformatics and genome-wide expression analyses with a new series of transgenic and cellular assays to show for the first time in malaria parasites that microarray read out from a chemical perturbation can have predictive value. We thereby identified and characterized an exported P. falciparum protein resident in a new vesicular compartment induced by the parasite in the erythrocyte. This protein, named Erythrocyte Vesicle Protein 1 (EVP1), shows novel dynamics of distribution in the parasite and intraerythrocytic membranes. Evidence is presented that its expression results in a change in TVN-mediated lipid import at the host membrane and that it is required for intracellular parasite growth, but not invasion. This exported protein appears to be needed for the maintenance of an essential tubovesicular nutrient import pathway induced by the pathogen in the host cell. Our approach may be generalized to the analysis of hundreds of 'hypothetical' P. falciparum proteins to understand their role in parasite entry and/or growth in erythrocytes as well as phenotypic contributions to either antigen export or tubovesicular import. By functionally validating these unknowns, one may identify new targets in host-microbial interactions for prophylaxis against this major human pathogen.


Subject(s)
Erythrocytes/metabolism , Intracellular Membranes/metabolism , Lipid Metabolism , Membrane Proteins/metabolism , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Animals , Biological Transport/genetics , Erythrocytes/parasitology , Gene Expression Regulation/genetics , Genome, Protozoan/genetics , Humans , Intracellular Membranes/parasitology , Lipid Metabolism/genetics , Lipids/genetics , Membrane Proteins/genetics , Plasmodium falciparum/genetics , Protozoan Proteins/genetics
17.
PLoS Pathog ; 4(6): e1000084, 2008 Jun 13.
Article in English | MEDLINE | ID: mdl-18551176

ABSTRACT

The malaria agent Plasmodium falciparum is predicted to export a "secretome" of several hundred proteins to remodel the host erythrocyte. Prediction of protein export is based on the presence of an ER-type signal sequence and a downstream Host-Targeting (HT) motif (which is similar to, but distinct from, the closely related Plasmodium Export Element [PEXEL]). Previous attempts to determine the entire secretome, using either the HT-motif or the PEXEL, have yielded large sets of proteins, which have not been comprehensively tested. We present here an expanded secretome that is optimized for both P. falciparum signal sequences and the HT-motif. From the most conservative of these three secretome predictions, we identify 11 proteins that are preserved across human- and rodent-infecting Plasmodium species. The conservation of these proteins likely indicates that they perform important functions in the interaction with and remodeling of the host erythrocyte important for all Plasmodium parasites. Using the piggyBac transposition system, we validate their export and find a positive prediction rate of approximately 70%. Even for proteins identified by all secretomes, the positive prediction rate is not likely to exceed approximately 75%. Attempted deletions of the genes encoding the conserved exported proteins were not successful, but additional functional analyses revealed the first conserved secretome function. This gave new insight into mechanisms for the assembly of the parasite-induced tubovesicular network needed for import of nutrients into the infected erythrocyte. Thus, genomic screens combined with functional assays provide unexpected and fundamental insights into host remodeling by this major human pathogen.


Subject(s)
Algorithms , Plasmodium falciparum/pathogenicity , Protein Sorting Signals , Protozoan Proteins/metabolism , Animals , Conserved Sequence , Erythrocytes/parasitology , Genomics/methods , Humans , Malaria , Plasmodium falciparum/chemistry , Protein Transport , Protozoan Proteins/genetics , Rodentia
18.
PLoS One ; 3(5): e2287, 2008 May 28.
Article in English | MEDLINE | ID: mdl-18509478

ABSTRACT

Malaria transmission depends on infective stages in the mosquito salivary glands. Plasmodium sporozoites that mature in midgut oocysts must traverse the hemocoel and invade the mosquito salivary glands in a process thought to be mediated by parasite ligands. MAEBL, a homologue of the transmembrane EBP ligands essential in merozoite invasion, is expressed abundantly in midgut sporozoites. Alternative splicing generates different MAEBL isoforms and so it is unclear what form is functionally essential. To identify the MAEBL isoform required for P. falciparum (NF54) sporozoite invasion of salivary glands, we created knockout and allelic replacements each carrying CDS of a single MAEBL isoform. Only the transmembrane form of MAEBL is essential and is the first P. falciparum ligand validated as essential for invasion of Anopheles salivary glands. MAEBL is the first P. falciparum ligand experimentally determined to be essential for this important step in the life cycle where the vector becomes infectious for transmitting sporozoites to people. With an increasing emphasis on advancing vector-based transgenic methods for suppression of malaria, it is important that this type of study, using modern molecular genetic tools, is done with the agent of the human disease. Understanding what P. falciparum sporozoite ligands are critical for mosquito transmission will help validate targets for vector-based transmission-blocking strategies.


Subject(s)
Anopheles/parasitology , Membrane Proteins/physiology , Plasmodium falciparum/pathogenicity , Protozoan Proteins/physiology , Receptors, Cell Surface/physiology , Salivary Glands/parasitology , Alleles , Animals , Base Sequence , DNA Primers , Female , Oocysts , Open Reading Frames , Protozoan Proteins/chemistry , Receptors, Cell Surface/chemistry , Reverse Transcriptase Polymerase Chain Reaction , Sporozoites
19.
Blood ; 111(4): 2418-26, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-18057226

ABSTRACT

In blood-stage infection by the human malaria parasite Plasmodium falciparum, export of proteins from the intracellular parasite to the erythrocyte is key to virulence. This export is mediated by a host-targeting (HT) signal present on a "secretome" of hundreds of parasite proteins engaged in remodeling the erythrocyte. However, the route of HT-mediated export is poorly understood. Here we show that minimal soluble and membrane protein reporters that contain the HT motif and mimic export of endogenous P falciparum proteins are detected in the lumen of "cleft" structures synthesized by the pathogen. Clefts are efficiently targeted by the HT signal. Furthermore, the HT signal does not directly translocate across the parasitophorous vacuolar membrane (PVM) surrounding the parasite to deliver protein to the erythrocyte cytoplasm, as suggested by current models of parasite protein trafficking to the erythrocyte. Rather, it is a lumenal signal that sorts protein into clefts, which then are exported beyond the PVM. These data suggest that Maurer's clefts, which are unique to the virulent P falciparum species, are pathogen-induced secretory organelles that concentrate HT-containing soluble and membrane parasite proteins in their lumen for delivery to the host erythrocyte.


Subject(s)
Erythrocytes/parasitology , Malaria, Falciparum/blood , Organelles/ultrastructure , Plasmodium falciparum/pathogenicity , Plasmodium falciparum/ultrastructure , Protozoan Proteins/metabolism , Animals , Base Sequence , Genes, Reporter , Humans , Intracellular Membranes/physiology , Molecular Sequence Data , Polymerase Chain Reaction , Protein Transport , Vacuoles/physiology , Virulence
20.
Int J Parasitol ; 37(1): 1-10, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17113093

ABSTRACT

Malaria is a global problem that affects millions of people annually. A relatively poor understanding of the malaria parasite biology has hindered vaccine and drug development against this disease. Robust methods for genetic analyses in Plasmodium have been lacking due to the difficulties in its genetic manipulation. Introduction of transfection technologies laid the foundation for genetic dissection of Plasmodium and recent years have seen the development of novel tools for genetic manipulation that will help us delineate the intriguing biology of this parasite. This review focuses on such recent advances in transfection technologies for Plasmodium that have improved our ability to carry out more thorough genetic analyses of the biology of the malaria parasite.


Subject(s)
Plasmodium/genetics , Transfection/methods , Animals , DNA Transposable Elements/genetics , DNA, Protozoan/genetics , Gene Expression/genetics , Genes, Protozoan/genetics , Genetic Markers/genetics , Humans , Malaria/genetics , Mutagenesis/genetics , Plasmodium berghei/genetics , Plasmodium falciparum/genetics , Transgenes/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...