Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Cell ; 171(7): 1532-1544.e15, 2017 Dec 14.
Article in English | MEDLINE | ID: mdl-29129376

ABSTRACT

Transmission represents a population bottleneck in the Plasmodium life cycle and a key intervention target of ongoing efforts to eradicate malaria. Sexual differentiation is essential for this process, as only sexual parasites, called gametocytes, are infective to the mosquito vector. Gametocyte production rates vary depending on environmental conditions, but external stimuli remain obscure. Here, we show that the host-derived lipid lysophosphatidylcholine (LysoPC) controls P. falciparum cell fate by repressing parasite sexual differentiation. We demonstrate that exogenous LysoPC drives biosynthesis of the essential membrane component phosphatidylcholine. LysoPC restriction induces a compensatory response, linking parasite metabolism to the activation of sexual-stage-specific transcription and gametocyte formation. Our results reveal that malaria parasites can sense and process host-derived physiological signals to regulate differentiation. These data close a critical knowledge gap in parasite biology and introduce a major component of the sexual differentiation pathway in Plasmodium that may provide new approaches for blocking malaria transmission.


Subject(s)
Lysophosphatidylcholines/metabolism , Malaria/parasitology , Plasmodium falciparum/growth & development , Plasmodium falciparum/metabolism , Animals , Female , Humans , Malaria/immunology , Metabolic Networks and Pathways , Mice , Mice, Inbred C57BL , Plasmodium berghei/physiology , Reproduction
2.
J Infect Dis ; 223(10): 1817-1821, 2021 05 28.
Article in English | MEDLINE | ID: mdl-32941614

ABSTRACT

Plasmodium vivax has 2 invasion ligand/host receptor pathways (P. vivax Duffy-binding protein/Duffy antigen receptor for chemokines [DARC] and P. vivax reticulocyte binding protein 2b/transferrin receptor [TfR1]) that are promising targets for therapeutic intervention. We optimized invasion assays with isogenic cultured reticulocytes. Using a receptor blockade approach with multiple P. vivax isolates, we found that all strains utilized both DARC and TfR1, but with significant variation in receptor usage. This suggests that P. vivax, like Plasmodium falciparum, uses alternative invasion pathways, with implications for pathogenesis and vaccine development.


Subject(s)
Antigens, CD , Duffy Blood-Group System , Malaria, Vivax , Plasmodium vivax , Receptors, Cell Surface , Receptors, Transferrin , Cells, Cultured , Humans , Plasmodium vivax/pathogenicity , Reticulocytes/parasitology
3.
Proc Natl Acad Sci U S A ; 114(44): E9356-E9365, 2017 10 31.
Article in English | MEDLINE | ID: mdl-29078358

ABSTRACT

During malaria blood-stage infections, Plasmodium parasites interact with the RBC surface to enable invasion followed by intracellular proliferation. Critical factors involved in invasion have been identified using biochemical and genetic approaches including specific knockdowns of genes of interest from primary CD34+ hematopoietic stem cells (cRBCs). Here we report the development of a robust in vitro culture system to produce RBCs that allow the generation of gene knockouts via CRISPR/Cas9 using the immortal JK-1 erythroleukemia line. JK-1 cells spontaneously differentiate, generating cells at different stages of erythropoiesis, including terminally differentiated nucleated RBCs that we term "jkRBCs." A screen of small-molecule epigenetic regulators identified several bromodomain-specific inhibitors that promote differentiation and enable production of synchronous populations of jkRBCs. Global surface proteomic profiling revealed that jkRBCs express all known Pfalciparum host receptors in a similar fashion to cRBCs and that multiple Pfalciparum strains invade jkRBCs at comparable levels to cRBCs and RBCs. Using CRISPR/Cas9, we deleted two host factors, basigin (BSG) and CD44, for which no natural nulls exist. BSG interacts with the parasite ligand Rh5, a prominent vaccine candidate. A BSG knockout was completely refractory to parasite invasion in a strain-transcendent manner, confirming the essential role for BSG during invasion. CD44 was recently identified in an RNAi screen of blood group genes as a host factor for invasion, and we show that CD44 knockout results in strain-transcendent reduction in invasion. Furthermore, we demonstrate a functional interaction between these two determinants in mediating Pfalciparum erythrocyte invasion.


Subject(s)
CRISPR-Cas Systems/genetics , Erythrocytes/metabolism , Erythrocytes/parasitology , Plasmodium falciparum/genetics , Antigens, Protozoan/metabolism , Basigin/metabolism , Carrier Proteins/metabolism , Cell Differentiation/physiology , Cell Line, Tumor , Clustered Regularly Interspaced Short Palindromic Repeats/physiology , Epigenesis, Genetic/physiology , Gene Knockout Techniques/methods , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/parasitology , Host-Parasite Interactions/physiology , Humans , Hyaluronan Receptors/metabolism , K562 Cells , Leukemia, Erythroblastic, Acute/metabolism , Leukemia, Erythroblastic, Acute/parasitology , Ligands , Malaria/parasitology , Malaria, Falciparum/metabolism , Malaria, Falciparum/parasitology , Proteomics/methods , Protozoan Proteins/metabolism
4.
Am J Hematol ; 94(9): 963-974, 2019 09.
Article in English | MEDLINE | ID: mdl-31148215

ABSTRACT

Malaria pathogenesis is caused by the replication of Plasmodium parasites within the red blood cells (RBCs) of the vertebrate host. This selective pressure has favored the evolution of protective polymorphisms in erythrocyte proteins, a subset of which serve as cognate receptors for parasite invasion ligands. Recently, the generation of RBCs from immortalized hematopoietic stem cells (HSCs) has offered a more tractable system for genetic manipulation and long-term in vitro culture, enabling elucidation of the functional determinants of host susceptibility in vitro. Here we report the generation of an immortalized erythroid progenitor cell line (EJ cells) from as few as 100 000 peripheral blood mononuclear cells. It offers a robust method for the creation of customized model systems from small volumes of peripheral blood. The EJ cell differentiation mirrored erythropoiesis of primary HSCs, yielding orthochromatic erythroblasts and enucleated RBCs after eight days (ejRBCs). The ejRBCs supported invasion by both P. vivax and P. falciparum. To demonstrate the genetic tractability of this system, we used CRISPR/Cas9 to disrupt the Duffy Antigen/Receptor for Chemokines (DARC) gene, which encodes the canonical receptor of P. vivax in humans. Invasion of P. vivax into this DARC-knockout cell line was strongly inhibited providing direct genetic evidence that P. vivax requires DARC for RBC invasion. Further, genetic complementation of DARC restored P. vivax invasion. Taken together, the peripheral blood immortalization method presented here offers the capacity to generate biologically representative model systems for studies of blood-stage malaria invasion from the peripheral blood of donors harboring unique genetic backgrounds, or rare polymorphisms.


Subject(s)
Erythroid Precursor Cells , Malaria, Falciparum , Malaria, Vivax , Models, Biological , Peripheral Blood Stem Cells , Plasmodium falciparum/metabolism , Plasmodium vivax/metabolism , Cell Line, Transformed , Erythroid Precursor Cells/metabolism , Erythroid Precursor Cells/parasitology , Erythroid Precursor Cells/physiology , Humans , Malaria, Falciparum/metabolism , Malaria, Falciparum/pathology , Malaria, Vivax/metabolism , Malaria, Vivax/pathology , Peripheral Blood Stem Cells/metabolism , Peripheral Blood Stem Cells/parasitology , Peripheral Blood Stem Cells/pathology
5.
Infect Immun ; 85(10)2017 10.
Article in English | MEDLINE | ID: mdl-28760933

ABSTRACT

Plasmodium falciparum, the parasite that causes the deadliest form of malaria, has evolved multiple proteins known as invasion ligands that bind to specific erythrocyte receptors to facilitate invasion of human erythrocytes. The EBA-175/glycophorin A (GPA) and Rh5/basigin ligand-receptor interactions, referred to as invasion pathways, have been the subject of intense study. In this study, we focused on the less-characterized sialic acid-containing receptors glycophorin B (GPB) and glycophorin C (GPC). Through bioinformatic analysis, we identified extensive variation in glycophorin B (GYPB) transcript levels in individuals from Benin, suggesting selection from malaria pressure. To elucidate the importance of the GPB and GPC receptors relative to the well-described EBA-175/GPA invasion pathway, we used an ex vivo erythrocyte culture system to decrease expression of GPA, GPB, or GPC via lentiviral short hairpin RNA transduction of erythroid progenitor cells, with global surface proteomic profiling. We assessed the efficiency of parasite invasion into knockdown cells using a panel of wild-type P. falciparum laboratory strains and invasion ligand knockout lines, as well as P. falciparum Senegalese clinical isolates and a short-term-culture-adapted strain. For this, we optimized an invasion assay suitable for use with small numbers of erythrocytes. We found that all laboratory strains and the majority of field strains tested were dependent on GPB expression level for invasion. The collective data suggest that the GPA and GPB receptors are of greater importance than the GPC receptor, supporting a hierarchy of erythrocyte receptor usage in P. falciparum.


Subject(s)
Erythrocytes/physiology , Erythrocytes/parasitology , Glycophorins/genetics , Plasmodium falciparum/pathogenicity , Computational Biology , Glycophorins/metabolism , Humans , Ligands , Plasmodium falciparum/immunology , Plasmodium falciparum/physiology , Protein Binding , Proteomics , Receptors, Cell Surface/metabolism
6.
PLoS Pathog ; 9(8): e1003546, 2013.
Article in English | MEDLINE | ID: mdl-23950716

ABSTRACT

Malaria blood stage parasites export a large number of proteins into their host erythrocyte to change it from a container of predominantly hemoglobin optimized for the transport of oxygen into a niche for parasite propagation. To understand this process, it is crucial to know which parasite proteins are exported into the host cell. This has been aided by the PEXEL/HT sequence, a five-residue motif found in many exported proteins, leading to the prediction of the exportome. However, several PEXEL/HT negative exported proteins (PNEPs) indicate that this exportome is incomplete and it remains unknown if and how many further PNEPs exist. Here we report the identification of new PNEPs in the most virulent malaria parasite Plasmodium falciparum. This includes proteins with a domain structure deviating from previously known PNEPs and indicates that PNEPs are not a rare exception. Unexpectedly, this included members of the MSP-7 related protein (MSRP) family, suggesting unanticipated functions of MSRPs. Analyzing regions mediating export of selected new PNEPs, we show that the first 20 amino acids of PNEPs without a classical N-terminal signal peptide are sufficient to promote export of a reporter, confirming the concept that this is a shared property of all PNEPs of this type. Moreover, we took advantage of newly found soluble PNEPs to show that this type of exported protein requires unfolding to move from the parasitophorous vacuole (PV) into the host cell. This indicates that soluble PNEPs, like PEXEL/HT proteins, are exported by translocation across the PV membrane (PVM), highlighting protein translocation in the parasite periphery as a general means in protein export of malaria parasites.


Subject(s)
Cell Membrane/metabolism , Plasmodium falciparum/metabolism , Protein Sorting Signals/physiology , Protozoan Proteins/metabolism , Animals , Cell Membrane/genetics , Mice , Plasmodium falciparum/genetics , Protein Transport/physiology , Protozoan Proteins/genetics
7.
Cell Microbiol ; 16(5): 612-20, 2014 May.
Article in English | MEDLINE | ID: mdl-24506567

ABSTRACT

Plasmodium knowlesi is a simian malaria parasite primarily infecting macaque species in Southeast Asia. Although its capacity to infect humans has been recognized since the early part of the last century, it has recently become evident that human infections are widespread and potentially life threatening. Historically, P. knowlesi has proven to be a powerful tool in early studies of malaria parasites, providing key breakthroughs in understanding many aspects of Plasmodium biology. However, the necessity to grow the parasite either in macaques or in vitro using macaque blood restricted research to laboratories with access to these resources. The recent adaptation of P. knowlesi to grow and proliferate in vitro in human red blood cells (RBCs) is therefore a substantial step towards revitalizing and expanding research on P. knowlesi. Furthermore, the development of a highly efficient transfection system to genetically modify the parasite makes P. knowlesi an ideal model to study parasite biology. In this review, we elaborate on the importance of P. knowlesi in earlier phases of malaria research and highlight the future potential of the newly available human adapted P. knowlesi parasite lines.


Subject(s)
Adaptation, Biological , Erythrocytes/parasitology , Host-Pathogen Interactions , Plasmodium knowlesi/physiology , Animals , History, 20th Century , History, 21st Century , Humans , Macaca , Parasitology/history , Parasitology/methods
8.
Cell Rep ; 43(4): 114012, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38573856

ABSTRACT

Plasmodium falciparum is a human-adapted apicomplexan parasite that causes the most dangerous form of malaria. P. falciparum cysteine-rich protective antigen (PfCyRPA) is an invasion complex protein essential for erythrocyte invasion. The precise role of PfCyRPA in this process has not been resolved. Here, we show that PfCyRPA is a lectin targeting glycans terminating with α2-6-linked N-acetylneuraminic acid (Neu5Ac). PfCyRPA has a >50-fold binding preference for human, α2-6-linked Neu5Ac over non-human, α2-6-linked N-glycolylneuraminic acid. PfCyRPA lectin sites were predicted by molecular modeling and validated by mutagenesis studies. Transgenic parasite lines expressing endogenous PfCyRPA with single amino acid exchange mutants indicated that the lectin activity of PfCyRPA has an important role in parasite invasion. Blocking PfCyRPA lectin activity with small molecules or with lectin-site-specific monoclonal antibodies can inhibit blood-stage parasite multiplication. Therefore, targeting PfCyRPA lectin activity with drugs, immunotherapy, or a vaccine-primed immune response is a promising strategy to prevent and treat malaria.


Subject(s)
Erythrocytes , Plasmodium falciparum , Polysaccharides , Protozoan Proteins , Humans , Antigens, Protozoan/metabolism , Antigens, Protozoan/immunology , Antigens, Protozoan/genetics , Erythrocytes/parasitology , Erythrocytes/metabolism , Lectins/metabolism , Lectins/genetics , Malaria, Falciparum/parasitology , Plasmodium falciparum/metabolism , Polysaccharides/metabolism , Protein Binding , Protozoan Proteins/metabolism , Protozoan Proteins/genetics
9.
Elife ; 122023 11 07.
Article in English | MEDLINE | ID: mdl-37934560

ABSTRACT

Plasmodium falciparum accounts for the majority of over 600,000 malaria-associated deaths annually. Parasites resistant to nearly all antimalarials have emerged and the need for drugs with alternative modes of action is thus undoubted. The FK506-binding protein PfFKBP35 has gained attention as a promising drug target due to its high affinity to the macrolide compound FK506 (tacrolimus). Whilst there is considerable interest in targeting PfFKBP35 with small molecules, a genetic validation of this factor as a drug target is missing and its function in parasite biology remains elusive. Here, we show that limiting PfFKBP35 levels are lethal to P. falciparum and result in a delayed death-like phenotype that is characterized by defective ribosome homeostasis and stalled protein synthesis. Our data furthermore suggest that FK506, unlike the action of this drug in model organisms, exerts its antiproliferative activity in a PfFKBP35-independent manner and, using cellular thermal shift assays, we identify putative FK506-targets beyond PfFKBP35. In addition to revealing first insights into the function of PfFKBP35, our results show that FKBP-binding drugs can adopt non-canonical modes of action - with major implications for the development of FK506-derived molecules active against Plasmodium parasites and other eukaryotic pathogens.


Subject(s)
Antimalarials , Malaria, Falciparum , Humans , Tacrolimus , Anti-Bacterial Agents , Drug Delivery Systems , Homeostasis , Tacrolimus Binding Proteins
10.
Mol Microbiol ; 71(4): 1003-17, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19170882

ABSTRACT

A short motif termed Plasmodium export element (PEXEL) or vacuolar targeting signal (VTS) characterizes Plasmodium proteins exported into the host cell. These proteins mediate host cell modifications essential for parasite survival and virulence. However, several PEXEL-negative exported proteins indicate that the currently predicted malaria exportome is not complete and it is unknown whether and how these proteins relate to PEXEL-positive export. Here we show that the N-terminal 10 amino acids of the PEXEL-negative exported protein REX2 (ring-exported protein 2) are necessary for its targeting and that a single-point mutation in this region abolishes export. Furthermore we show that the REX2 transmembrane domain is also essential for export and that together with the N-terminal region it is sufficient to promote export of another protein. An N-terminal region and the transmembrane domain of the unrelated PEXEL-negative exported protein SBP1 (skeleton-binding protein 1) can functionally replace the corresponding regions in REX2, suggesting that these sequence features are also present in other PEXEL-negative exported proteins. Similar to PEXEL proteins we find that REX2 is processed, but in contrast, detect no evidence for N-terminal acetylation.


Subject(s)
Membrane Proteins/metabolism , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Amino Acid Sequence , Animals , Erythrocytes/parasitology , Membrane Proteins/genetics , Molecular Sequence Data , Plasmodium falciparum/genetics , Point Mutation , Protein Transport , Protozoan Proteins/genetics , Sequence Alignment , Sequence Deletion
11.
Trends Parasitol ; 36(2): 85-87, 2020 02.
Article in English | MEDLINE | ID: mdl-31883707

ABSTRACT

A recent report by Jennison et al. reveals an important role for plasmepsin V (PMV), an aspartyl protease, in the development of malaria transmission stages. The authors showed that PMV activity is critical for protein export in these stages and that specific PMV inhibitors block parasite transmission to mosquitoes.


Subject(s)
Aspartic Acid Endopeptidases/antagonists & inhibitors , Aspartic Acid Endopeptidases/metabolism , Malaria/parasitology , Malaria/transmission , Plasmodium/enzymology , Animals , Antimalarials/pharmacology , Carbamates/pharmacology , Humans , Life Cycle Stages/drug effects , Life Cycle Stages/physiology , Malaria/prevention & control , Oligopeptides/pharmacology , Plasmodium/drug effects , Protein Transport/drug effects
12.
Nat Commun ; 7: 12727, 2016 10 10.
Article in English | MEDLINE | ID: mdl-27721445

ABSTRACT

Malaria remains one of the greatest public health challenges worldwide, particularly in sub-Saharan Africa. The clinical outcome of individuals infected with Plasmodium falciparum parasites depends on many factors including host systemic inflammatory responses, parasite sequestration in tissues and vascular dysfunction. Production of pro-inflammatory cytokines and chemokines promotes endothelial activation as well as recruitment and infiltration of inflammatory cells, which in turn triggers further endothelial cell activation and parasite sequestration. Inflammatory responses are triggered in part by bioactive parasite products such as hemozoin and infected red blood cell-derived extracellular vesicles (iRBC-derived EVs). Here we demonstrate that such EVs contain functional miRNA-Argonaute 2 complexes that are derived from the host RBC. Moreover, we show that EVs are efficiently internalized by endothelial cells, where the miRNA-Argonaute 2 complexes modulate target gene expression and barrier properties. Altogether, these findings provide a mechanistic link between EVs and vascular dysfunction during malaria infection.


Subject(s)
Argonaute Proteins/metabolism , Blood Vessels/metabolism , Erythrocytes/parasitology , Extracellular Vesicles/metabolism , Malaria, Falciparum/metabolism , Malaria, Falciparum/parasitology , MicroRNAs/metabolism , Brain/blood supply , Cell Line , Endocytosis , Endothelial Cells/metabolism , Erythrocytes/ultrastructure , Extracellular Vesicles/ultrastructure , Gene Expression Regulation , Gene Silencing , Humans , MicroRNAs/genetics , Microvessels/cytology , RNA-Induced Silencing Complex/metabolism
13.
Methods Enzymol ; 506: 81-92, 2012.
Article in English | MEDLINE | ID: mdl-22341220

ABSTRACT

Life cell imaging is a tool for cell biology that has provided invaluable insights into many dynamic processes such as cell division, morphogenesis, or endo- and exocytosis. While observing cells by time-lapse imaging is a standard procedure in many systems, this technique was until recently not available for blood stages of Plasmodium falciparum, the causative agent of the most severe form of human malaria. Here, we provide a detailed description of the procedure for time-lapse-based four-dimensional microscopy in blood stages of this important pathogen. With the widespread use of P. falciparum transfection to fluorescently tag proteins of interest, this technique provides a new tool to study the biology of malaria blood stages that is hoped to lead to a better appreciation of the dynamic processes in this life cycle phase.


Subject(s)
Malaria, Falciparum/parasitology , Microscopy, Confocal/methods , Microscopy, Fluorescence/methods , Plasmodium falciparum/growth & development , Plasmodium falciparum/ultrastructure , Staining and Labeling/methods , Animals , Fluorescent Dyes/analysis , Humans , Malaria, Falciparum/blood
14.
Cell Host Microbe ; 12(5): 717-29, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-23159060

ABSTRACT

For proliferation, the malaria parasite Plasmodium falciparum needs to modify the infected host cell extensively. To achieve this, the parasite exports proteins containing a Plasmodium export element (PEXEL) into the host cell. Phosphatidylinositol-3-phosphate binding and cleavage of the PEXEL are thought to mediate protein export. We show that these requirements can be bypassed, exposing a second level of export control in the N terminus generated after PEXEL cleavage that is sufficient to distinguish exported from nonexported proteins. Furthermore, this region also corresponds to the export domain of a second group of exported proteins lacking PEXELs (PNEPs), indicating shared export properties among different exported parasite proteins. Concordantly, export of both PNEPs and PEXEL proteins depends on unfolding, revealing translocation as a common step in export. However, translocation of transmembrane proteins occurs at the parasite plasma membrane, one step before translocation of soluble proteins, indicating unexpectedly complex translocation events at the parasite periphery.


Subject(s)
Carrier Proteins/metabolism , Malaria, Falciparum/metabolism , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Animals , Aspartic Acid Endopeptidases/metabolism , Humans , Phosphatidylinositol Phosphates/metabolism , Protein Transport , Protein Unfolding , Protozoan Proteins/chemistry
15.
Nat Commun ; 2: 165, 2011 Jan 25.
Article in English | MEDLINE | ID: mdl-21266965

ABSTRACT

Blood stages of Plasmodium falciparum cause the pathology of malaria; however, the progression of the parasite through this complex part of the life cycle has never been visualized. In this study, we use four-dimensional imaging to show for the first time the development of individual parasites in erythrocytes and the concomitant host cell modifications. Our data visualize an unexpectedly dynamic parasite, provide a reference for this life cycle stage and challenge the model that protein export in P. falciparum is linked to the biogenesis of host cell modifications termed Maurer's clefts. Our results provide a novel view of the blood-stage development, Maurer's cleft development and protein export in malaria parasites, and open the door to study dynamic processes, drug effects and the phenotype of mutants.

SELECTION OF CITATIONS
SEARCH DETAIL