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










Publication year range
1.
Immunol Cell Biol ; 101(9): 857-866, 2023 10.
Article in English | MEDLINE | ID: mdl-37593973

ABSTRACT

Current serological tests cannot differentiate between total immunoglobulin A (IgA) and dimeric IgA (dIgA) associated with mucosal immunity. Here, we describe two new assays, dIgA-ELISA and dIgA-multiplex bead assay (MBA), that utilize the preferential binding of dIgA to a chimeric form of secretory component, allowing the differentiation between dIgA and monomeric IgA. dIgA responses elicited through severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection were measured in (i) a longitudinal panel, consisting of 74 samples (n = 20 individuals) from hospitalized cases of coronavirus disease 2019 (COVID-19); (ii) a longitudinal panel, consisting of 96 samples (n = 10 individuals) from individuals with mild COVID-19; (iii) a cross-sectional panel with PCR-confirmed SARS-CoV-2 infection with mild COVID-19 (n = 199) and (iv) pre-COVID-19 samples (n = 200). The dIgA-ELISA and dIgA-MBA demonstrated a specificity for dIgA of 99% and 98.5%, respectively. Analysis of dIgA responses in the longitudinal panels revealed that 70% (ELISA) and 50% (MBA) of patients elicited a dIgA response by day 20 after PCR diagnosis with a SARS-CoV-2 infection. Individuals with mild COVID-19 displayed increased levels of dIgA within the first 3 weeks after diagnosis but responses appeared to be short lived, compared with sustained IgA levels. However, in samples from hospitalized patients with COVID-19 we observed high and sustained levels of dIgA, up to 245 days after PCR diagnosis. Our results suggest that severe COVID-19 infections are associated with sustained levels of plasma dIgA compared with mild cases.


Subject(s)
COVID-19 , Humans , COVID-19/diagnosis , SARS-CoV-2/metabolism , Cross-Sectional Studies , Immunoglobulin A , Antibodies, Viral , Immunoglobulin M
2.
Trends Parasitol ; 39(8): 622-625, 2023 08.
Article in English | MEDLINE | ID: mdl-37302957

ABSTRACT

Protein disulfide isomerases (PDIs) ensure that specific substrate proteins are correctly folded. PDI activity plays an essential role in malaria transmission. Here we provide an overview of the role of PDIs in malaria-causing Plasmodium parasites and outline why PDI inhibition could be a novel way to treat malaria and prevent transmission.


Subject(s)
Malaria , Plasmodium , Humans , Protein Disulfide-Isomerases/metabolism , Malaria/drug therapy , Malaria/prevention & control , Plasmodium/metabolism
3.
Parasitol Int ; 87: 102526, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34896312

ABSTRACT

Plasmodium vivax is the most geographically widespread human malaria parasite. Global malaria efforts have been less successful at reducing the burden of P. vivax compared to P. falciparum, owing to the unique biology and related treatment complexity of P. vivax. As a result, P. vivax is now the dominant malaria parasite throughout the Asia-Pacific and South America causing up to 14 million clinical cases every year and is considered a major obstacle to malaria elimination. Key features circumventing existing malaria control tools are the transmissibility of asymptomatic, low-density circulating infections and reservoirs of persistent dormant liver stages (hypnozoites) that are undetectable but reactivate to cause relapsing infections and sustain transmission. In this review we summarise the new knowledge shaping our understanding of the global epidemiology of P. vivax infections, highlighting the challenges for elimination and the tools that will be required achieve this.


Subject(s)
Disease Reservoirs/parasitology , Malaria, Vivax , Plasmodium vivax/physiology , Humans , Liver/parasitology , Malaria, Falciparum/epidemiology , Malaria, Falciparum/prevention & control , Malaria, Vivax/epidemiology , Malaria, Vivax/prevention & control , Malaria, Vivax/transmission , Plasmodium vivax/isolation & purification
4.
Elife ; 102021 12 23.
Article in English | MEDLINE | ID: mdl-34939934

ABSTRACT

HAP2 is a transmembrane gamete fusogen found in multiple eukaryotic kingdoms and is structurally homologous to viral class II fusogens. Studies in Plasmodium have suggested that HAP2 is an attractive target for vaccines that block transmission of malaria. HAP2 has three extracellular domains, arranged in the order D2, D1, and D3. Here, we report monoclonal antibodies against the D3 fragment of Plasmodium berghei HAP2 and crystal structures of D3 in complex with Fab fragments of two of these antibodies, one of which blocks fertilization of Plasmodium berghei in vitro and transmission of malaria in mosquitoes. We also show how this Fab binds the complete HAP2 ectodomain with electron microscopy. The two antibodies cross-react with HAP2 among multiple plasmodial species. Our characterization of the Plasmodium D3 structure, HAP2 ectodomain architecture, and mechanism of inhibition provide insights for the development of a vaccine to block malaria transmission.


Subject(s)
Antibodies, Monoclonal/metabolism , Germ Cells/immunology , Malaria/prevention & control , Malaria/transmission , Plasmodium berghei/immunology , Protozoan Proteins/immunology , Protozoan Proteins/metabolism , Animals , Binding Sites, Antibody , Biophysical Phenomena , Culicidae/parasitology , Germ Cells/physiology , Malaria/immunology , Membrane Fusion , Protein Binding , Protozoan Proteins/chemistry
5.
Methods Protoc ; 4(4)2021 Oct 09.
Article in English | MEDLINE | ID: mdl-34698238

ABSTRACT

Serology tests are extremely useful for assessing whether a person has been infected with a pathogen. Since the onset of the COVID-19 pandemic, measurement of anti-SARS-CoV-2-specific antibodies has been considered an essential tool in identifying seropositive individuals and thereby understanding the extent of transmission in communities. The Luminex system is a bead-based technology that has the capacity to assess multiple antigens simultaneously using very low sample volumes and is ideal for high-throughput studies. We have adapted this technology to develop a COVID-19 multi-antigen serological assay. This protocol described here carefully outlines recommended steps to optimize and establish this method for COVID-19-specific antibody measurement in plasma and in saliva. However, the protocol can easily be customized and thus the assay is broadly applicable to measure antibodies to other pathogens.

6.
Sci Rep ; 11(1): 1888, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33479319

ABSTRACT

New antimalarial therapeutics are needed to ensure that malaria cases continue to be driven down, as both emerging parasite resistance to frontline chemotherapies and mosquito resistance to current insecticides threaten control programmes. Plasmodium, the apicomplexan parasite responsible for malaria, causes disease pathology through repeated cycles of invasion and replication within host erythrocytes (the asexual cycle). Antimalarial drugs primarily target this cycle, seeking to reduce parasite burden within the host as fast as possible and to supress recrudescence for as long as possible. Intense phenotypic drug screening efforts have identified a number of promising new antimalarial molecules. Particularly important is the identification of compounds with new modes of action within the parasite to combat existing drug resistance and suitable for formulation of efficacious combination therapies. Here we detail the antimalarial properties of DDD01034957-a novel antimalarial molecule which is fast-acting and potent against drug resistant strains in vitro, shows activity in vivo, and possesses a resistance mechanism linked to the membrane transporter PfABCI3. These data support further medicinal chemistry lead-optimization of DDD01034957 as a novel antimalarial chemical class and provide new insights to further reduce in vivo metabolic clearance.


Subject(s)
Antimalarials/pharmacology , Drug Resistance/drug effects , Malaria/drug therapy , Plasmodium falciparum/drug effects , Animals , Antimalarials/chemistry , Erythrocytes/parasitology , Host-Parasite Interactions/drug effects , Humans , Inhibitory Concentration 50 , Malaria/parasitology , Mice , Molecular Structure , Plasmodium/drug effects , Plasmodium/parasitology , Plasmodium berghei/drug effects , Plasmodium berghei/parasitology , Plasmodium falciparum/physiology , Species Specificity
7.
Proc Natl Acad Sci U S A ; 117(13): 7363-7373, 2020 03 31.
Article in English | MEDLINE | ID: mdl-32165544

ABSTRACT

After being ingested by a female Anopheles mosquito during a bloodmeal on an infected host, and before they can reach the mosquito salivary glands to be transmitted to a new host, Plasmodium parasites must establish an infection of the mosquito midgut in the form of oocysts. To achieve this, they must first survive a series of robust innate immune responses that take place prior to, during, and immediately after ookinete traversal of the midgut epithelium. Understanding how parasites may evade these responses could highlight new ways to block malaria transmission. We show that an ookinete and sporozoite surface protein designated as PIMMS43 (Plasmodium Infection of the Mosquito Midgut Screen 43) is required for parasite evasion of the Anopheles coluzzii complement-like response. Disruption of PIMMS43 in the rodent malaria parasite Plasmodium berghei triggers robust complement activation and ookinete elimination upon mosquito midgut traversal. Silencing components of the complement-like system through RNAi largely restores ookinete-to-oocyst transition but oocysts remain small in size and produce a very small number of sporozoites that additionally are not infectious, indicating that PIMMS43 is also essential for sporogonic development in the oocyst. Antibodies that bind PIMMS43 interfere with parasite immune evasion when ingested with the infectious blood meal and significantly reduce the prevalence and intensity of infection. PIMMS43 genetic structure across African Plasmodium falciparum populations indicates allelic adaptation to sympatric vector populations. These data add to our understanding of mosquito-parasite interactions and identify PIMMS43 as a target of malaria transmission blocking.


Subject(s)
Anopheles/immunology , Mosquito Vectors/immunology , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Animals , Anopheles/metabolism , Anopheles/parasitology , Female , Host-Parasite Interactions/immunology , Humans , Immune Evasion , Malaria, Falciparum/parasitology , Malaria, Falciparum/transmission , Mosquito Vectors/metabolism , Mosquito Vectors/parasitology , Oocysts/immunology , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Sporozoites/immunology
8.
Malar J ; 18(1): 381, 2019 Nov 29.
Article in English | MEDLINE | ID: mdl-31783858

ABSTRACT

BACKGROUND: Malaria disease commences when blood-stage parasites, called merozoites, invade human erythrocytes. Whilst the process of invasion is traditionally seen as being entirely merozoite-driven, emerging data suggests erythrocyte biophysical properties markedly influence invasion. Cholesterol is a major determinant of cell membrane biophysical properties demanding its interrogation as a potential mediator of resistance to merozoite invasion of the erythrocyte. METHODS: Biophysical measurements of erythrocyte deformability by flicker spectroscopy were used to assess changes in erythrocyte bending modulus on forced integration of cholesterol and how these artificial changes affect invasion by human Plasmodium falciparum merozoites. To validate these observations in a natural context, either murine Plasmodium berghei or human Plasmodium falciparum merozoites were tested for their ability to invade erythrocytes from a hypercholesterolaemic mouse model or human clinical erythrocyte samples deriving from patients with a range of serum cholesterol concentrations, respectively. RESULTS: Erythrocyte bending modulus (a measure of deformability) was shown to be markedly affected by artificial modulation of cholesterol content and negatively correlated with merozoite invasion efficiency. In an in vitro infection context, however, erythrocytes taken from hypercholesterolaemic mice or from human clinical samples with varying serum cholesterol levels showed little difference in their susceptibility to merozoite invasion. Explaining this, membrane cholesterol levels in both mouse and human hypercholesterolaemia erythrocytes were subsequently found to be no different from matched normal serum controls. CONCLUSIONS: Based on these observations, serum cholesterol does not appear to impact on erythrocyte susceptibility to merozoite entry. Indeed, no relationship between serum cholesterol and cholesterol content of the erythrocyte is apparent. This work, nonetheless, suggests that native polymorphisms which do affect membrane lipid composition would be expected to affect parasite entry. This supports investigation of erythrocyte biophysical properties in endemic settings, which may yet identify naturally protective lipid-related polymorphisms.


Subject(s)
Cholesterol/blood , Dyslipidemias/etiology , Erythrocytes/parasitology , Malaria/physiopathology , Plasmodium berghei/physiology , Plasmodium falciparum/physiology , Animals , Biophysical Phenomena , Humans , Malaria, Falciparum/physiopathology , Male , Mice
9.
Sci Rep ; 9(1): 18300, 2019 12 04.
Article in English | MEDLINE | ID: mdl-31797966

ABSTRACT

Inhibiting transmission of Plasmodium is an essential strategy in malaria eradication, and the biological process of gamete fusion during fertilization is a proven target for this approach. Lack of knowledge of the mechanisms underlying fertilization have been a hindrance in the development of transmission-blocking interventions. Here we describe a protein disulphide isomerase essential for malarial transmission (PDI-Trans/PBANKA_0820300) to the mosquito. We show that PDI-Trans activity is male-specific, surface-expressed, essential for fertilization/transmission, and exhibits disulphide isomerase activity which is up-regulated post-gamete activation. We demonstrate that PDI-Trans is a viable anti-malarial drug and vaccine target blocking malarial transmission with the use of PDI inhibitor bacitracin (98.21%/92.48% reduction in intensity/prevalence), and anti-PDI-Trans antibodies (66.22%/33.16% reduction in intensity/prevalence). To our knowledge, these results provide the first evidence that PDI function is essential for malarial transmission, and emphasize the potential of anti-PDI agents to act as anti-malarials, facilitating the future development of novel transmission-blocking interventions.


Subject(s)
Antimalarials , Bacitracin , Malaria Vaccines , Malaria , Plasmodium berghei/enzymology , Protein Disulfide-Isomerases/physiology , Animals , Antimalarials/pharmacology , Antimalarials/therapeutic use , Bacitracin/pharmacology , Bacitracin/therapeutic use , Female , Malaria/prevention & control , Malaria/transmission , Malaria Vaccines/pharmacology , Malaria Vaccines/therapeutic use , Male , Mice , Plasmodium berghei/drug effects , Plasmodium berghei/pathogenicity , Protozoan Proteins/physiology
10.
Nat Commun ; 9(1): 4248, 2018 10 12.
Article in English | MEDLINE | ID: mdl-30315162

ABSTRACT

In malaria parasites, evolution of parasitism has been linked to functional optimisation. Despite this optimisation, most members of a calcium-dependent protein kinase (CDPK) family show genetic redundancy during erythrocytic proliferation. To identify relationships between phospho-signalling pathways, we here screen 294 genetic interactions among protein kinases in Plasmodium berghei. This reveals a synthetic negative interaction between a hypomorphic allele of the protein kinase G (PKG) and CDPK4 to control erythrocyte invasion which is conserved in P. falciparum. CDPK4 becomes critical when PKG-dependent calcium signals are attenuated to phosphorylate proteins important for the stability of the inner membrane complex, which serves as an anchor for the acto-myosin motor required for motility and invasion. Finally, we show that multiple kinases functionally complement CDPK4 during erythrocytic proliferation and transmission to the mosquito. This study reveals how CDPKs are wired within a stage-transcending signalling network to control motility and host cell invasion in malaria parasites.


Subject(s)
Epistasis, Genetic/genetics , Plasmodium berghei/metabolism , Plasmodium berghei/pathogenicity , Plasmodium falciparum/metabolism , Plasmodium falciparum/pathogenicity , Protein Kinases/metabolism , Protozoan Proteins/metabolism , Animals , Calcium/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Female , Malaria, Falciparum/parasitology , Male , Mice , Protein Kinases/genetics , Protozoan Proteins/genetics
11.
Elife ; 72018 06 19.
Article in English | MEDLINE | ID: mdl-29914622

ABSTRACT

Anti-malarial pre-erythrocytic vaccines (PEV) target transmission by inhibiting human infection but are currently partially protective. It has been posited, but never demonstrated, that co-administering transmission-blocking vaccines (TBV) would enhance malaria control. We hypothesized a mechanism that TBV could reduce parasite density in the mosquito salivary glands, thereby enhancing PEV efficacy. This was tested using a multigenerational population assay, passaging Plasmodium berghei to Anopheles stephensi mosquitoes. A combined efficacy of 90.8% (86.7-94.2%) was observed in the PEV +TBV antibody group, higher than the estimated efficacy of 83.3% (95% CrI 79.1-87.0%) if the two antibodies acted independently. Higher PEV efficacy at lower mosquito parasite loads was observed, comprising the first direct evidence that co-administering anti-sporozoite and anti-transmission interventions act synergistically, enhancing PEV efficacy across a range of TBV doses and transmission intensities. Combining partially effective vaccines of differing anti-parasitic classes is a pragmatic, powerful way to accelerate malaria elimination efforts.


Subject(s)
Antibodies, Blocking/administration & dosage , Antibodies, Monoclonal/administration & dosage , Antibodies, Protozoan/administration & dosage , Malaria Vaccines/administration & dosage , Malaria/prevention & control , Plasmodium berghei/immunology , Sporozoites/immunology , Animals , Anopheles/parasitology , Drug Synergism , Female , Humans , Malaria/immunology , Malaria/parasitology , Mice , Mosquito Vectors/parasitology , Parasite Load , Plasmodium berghei/drug effects , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Salivary Glands/parasitology , Sporozoites/chemistry , Trophozoites/chemistry , Trophozoites/immunology
13.
Cell Rep ; 21(10): 2868-2878, 2017 Dec 05.
Article in English | MEDLINE | ID: mdl-29212032

ABSTRACT

Inhibiting transmission of Plasmodium is a central strategy in malarial eradication, and the biological process of gamete fusion during fertilization is a proven target for this approach. The lack of a structure or known molecular function of current anti-malarial vaccine targets has previously been a hindrance in the development of transmission-blocking vaccines. Structure/function studies have indicated that the conserved gamete membrane fusion protein HAP2 is a class II viral fusion protein. Here, we demonstrate that targeting a function-critical site of the fusion/cd loop with species-specific antibodies reduces Plasmodium berghei transmission in vivo by 58.9% and in vitro fertilization by up to 89.9%. A corresponding reduction in P. falciparum transmission (75.5%/36.4% reductions in intensity/prevalence) is observed in complimentary field studies. These results emphasize conserved mechanisms of fusion in Apicomplexa, while highlighting an approach to design future anti-malarial transmission-blocking vaccines.


Subject(s)
Antimalarials/pharmacology , Malaria, Falciparum/drug therapy , Plasmodium berghei/drug effects , Protozoan Proteins/metabolism , Animals , Antimalarials/therapeutic use , Malaria/drug therapy , Malaria/genetics , Malaria/metabolism , Malaria Vaccines/therapeutic use , Malaria, Falciparum/genetics , Malaria, Falciparum/metabolism , Male , Mice , Plasmodium berghei/metabolism , Protozoan Proteins/genetics
14.
PLoS Pathog ; 13(1): e1006108, 2017 01.
Article in English | MEDLINE | ID: mdl-28081253

ABSTRACT

Over a century since Ronald Ross discovered that malaria is caused by the bite of an infectious mosquito it is still unclear how the number of parasites injected influences disease transmission. Currently it is assumed that all mosquitoes with salivary gland sporozoites are equally infectious irrespective of the number of parasites they harbour, though this has never been rigorously tested. Here we analyse >1000 experimental infections of humans and mice and demonstrate a dose-dependency for probability of infection and the length of the host pre-patent period. Mosquitoes with a higher numbers of sporozoites in their salivary glands following blood-feeding are more likely to have caused infection (and have done so quicker) than mosquitoes with fewer parasites. A similar dose response for the probability of infection was seen for humans given a pre-erythrocytic vaccine candidate targeting circumsporozoite protein (CSP), and in mice with and without transfusion of anti-CSP antibodies. These interventions prevented infection more efficiently from bites made by mosquitoes with fewer parasites. The importance of parasite number has widespread implications across malariology, ranging from our basic understanding of the parasite, how vaccines are evaluated and the way in which transmission should be measured in the field. It also provides direct evidence for why the only registered malaria vaccine RTS,S was partially effective in recent clinical trials.


Subject(s)
Anopheles/parasitology , Insect Vectors/parasitology , Malaria Vaccines/administration & dosage , Malaria/prevention & control , Plasmodium/immunology , Animals , Antibodies, Protozoan , Disease Models, Animal , Humans , Malaria/parasitology , Malaria/transmission , Mice , Plasmodium/growth & development , Population Dynamics , Protozoan Proteins/immunology , Salivary Glands/parasitology , Sporozoites/immunology , Vaccination
15.
Sci Rep ; 6: 19766, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26830761

ABSTRACT

The invasive blood-stage malaria parasite - the merozoite - induces rapid morphological changes to the target erythrocyte during entry. However, evidence for active molecular changes in the host cell that accompany merozoite invasion is lacking. Here, we use invasion inhibition assays, erythrocyte resealing and high-definition imaging to explore red cell responses during invasion. We show that although merozoite entry does not involve erythrocyte actin reorganisation, it does require ATP to complete the process. Towards dissecting the ATP requirement, we present an in depth quantitative phospho-proteomic analysis of the erythrocyte during each stage of invasion. Specifically, we demonstrate extensive increased phosphorylation of erythrocyte proteins on merozoite attachment, including modification of the cytoskeletal proteins beta-spectrin and PIEZO1. The association with merozoite contact but not active entry demonstrates that parasite-dependent phosphorylation is mediated by host-cell kinase activity. This provides the first evidence that the erythrocyte is stimulated to respond to early invasion events through molecular changes in its membrane architecture.


Subject(s)
Cytoskeleton/metabolism , Erythrocytes/metabolism , Merozoites/metabolism , Phosphoproteins/metabolism , Plasmodium falciparum/metabolism , Protein Kinases/metabolism , Erythrocytes/parasitology , Humans , Ion Channels/metabolism , Phosphorylation , Proteomics , Spectrin/metabolism
16.
BMC Biol ; 13: 52, 2015 Jul 18.
Article in English | MEDLINE | ID: mdl-26187647

ABSTRACT

BACKGROUND: Malaria invasion of red blood cells involves multiple parasite-specific targets that are easily accessible to inhibitory compounds, making it an attractive target for antimalarial development. However, no current antimalarial agents act against host cell invasion. RESULTS: Here, we demonstrate that the clinically used macrolide antibiotic azithromycin, which is known to kill human malaria asexual blood-stage parasites by blocking protein synthesis in their apicoplast, is also a rapid inhibitor of red blood cell invasion in human (Plasmodium falciparum) and rodent (P. berghei) malarias. Multiple lines of evidence demonstrate that the action of azithromycin in inhibiting parasite invasion of red blood cells is independent of its inhibition of protein synthesis in the parasite apicoplast, opening up a new strategy to develop a single drug with multiple parasite targets. We identified derivatives of azithromycin and erythromycin that are better invasion inhibitors than parent compounds, offering promise for development of this novel antimalarial strategy. CONCLUSIONS: Safe and effective macrolide antibiotics with dual modalities could be developed to combat malaria and reduce the parasite's options for resistance.


Subject(s)
Antimalarials/pharmacology , Azithromycin/pharmacology , Erythrocytes/parasitology , Erythromycin/pharmacology , Malaria/drug therapy , Plasmodium berghei/drug effects , Plasmodium falciparum/drug effects , Animals , Anopheles , Antimalarials/chemistry , Azithromycin/chemistry , Erythromycin/chemistry , Host-Parasite Interactions/drug effects , Humans , Malaria/parasitology , Mice , Plasmodium berghei/physiology , Plasmodium falciparum/physiology
17.
Malar J ; 14: 280, 2015 Jul 18.
Article in English | MEDLINE | ID: mdl-26187846

ABSTRACT

BACKGROUND: Gliding motility in Plasmodium parasites, the aetiological agents of malaria disease, is mediated by an actomyosin motor anchored in the outer pellicle of the motile cell. Effective motility is dependent on a parasite myosin motor and turnover of dynamic parasite actin filaments. To date, however, the basis for directional motility is not known. Whilst myosin is very likely orientated as a result of its anchorage within the parasite, how actin filaments are orientated to facilitate directional force generation remains unexplained. In addition, recent evidence has questioned the linkage between actin filaments and secreted surface antigens leaving the way by which motor force is transmitted to the extracellular milieu unknown. Malaria parasites possess a markedly reduced repertoire of actin regulators, among which few are predicted to interact with filamentous (F)-actin directly. One of these, PF3D7_1251200, shows strong homology to the coronin family of actin-filament binding proteins, herein referred to as PfCoronin. METHODS: Here the N terminal beta propeller domain of PfCoronin (PfCor-N) was expressed to assess its ability to bind and bundle pre-formed actin filaments by sedimentation assay, total internal reflection fluorescence (TIRF) microscopy and confocal imaging as well as to explore its ability to bind phospholipids. In parallel a tagged PfCoronin line in Plasmodium falciparum was generated to determine the cellular localization of the protein during asexual parasite development and blood-stage merozoite invasion. RESULTS: A combination of biochemical approaches demonstrated that the N-terminal beta-propeller domain of PfCoronin is capable of binding F-actin and facilitating formation of parallel filament bundles. In parasites, PfCoronin is expressed late in the asexual lifecycle and localizes to the pellicle region of invasive merozoites before and during erythrocyte entry. PfCoronin also associates strongly with membranes within the cell, likely mediated by interactions with phosphatidylinositol-4,5-bisphosphate (PI(4,5)P2) at the plasma membrane. CONCLUSIONS: These data suggest PfCoronin may fulfil a key role as the critical determinant of actin filament organization in the Plasmodium cell. This raises the possibility that macro-molecular organization of actin mediates directional motility in gliding parasites.


Subject(s)
Actin Cytoskeleton/chemistry , Microfilament Proteins/chemistry , Plasmodium falciparum/chemistry , Plasmodium falciparum/physiology , Protozoan Proteins/chemistry , Actin Cytoskeleton/metabolism , Animals , Erythrocytes/parasitology , Humans , Malaria, Falciparum/parasitology , Microfilament Proteins/metabolism , Models, Molecular , Plasmodium falciparum/metabolism , Plasmodium falciparum/pathogenicity , Protozoan Proteins/metabolism , Rabbits
18.
Cell Microbiol ; 16(5): 734-50, 2014 May.
Article in English | MEDLINE | ID: mdl-24612056

ABSTRACT

Motility is a fundamental part of cellular life and survival, including for Plasmodium parasites--single-celled protozoan pathogens responsible for human malaria. The motile life cycle forms achieve motility, called gliding, via the activity of an internal actomyosin motor. Although gliding is based on the well-studied system of actin and myosin, its core biomechanics are not completely understood. Currently accepted models suggest it results from a specifically organized cellular motor that produces a rearward directional force. When linked to surface-bound adhesins, this force is passaged to the cell posterior, propelling the parasite forwards. Gliding motility is observed in all three life cycle stages of Plasmodium: sporozoites, merozoites and ookinetes. However, it is only the ookinetes--formed inside the midgut of infected mosquitoes--that display continuous gliding without the necessity of host cell entry. This makes them ideal candidates for invasion-free biomechanical analysis. Here we apply a plate-based imaging approach to study ookinete motion in three-dimensional (3D) space to understand Plasmodium cell motility and how movement facilitates midgut colonization. Using single-cell tracking and numerical analysis of parasite motion in 3D, our analysis demonstrates that ookinetes move with a conserved left-handed helical trajectory. Investigation of cell morphology suggests this trajectory may be based on the ookinete subpellicular cytoskeleton, with complementary whole and subcellular electron microscopy showing that, like their motion paths, ookinetes share a conserved left-handed corkscrew shape and underlying twisted microtubular architecture. Through comparisons of 3D movement between wild-type ookinetes and a cytoskeleton-knockout mutant we demonstrate that perturbation of cell shape changes motion from helical to broadly linear. Therefore, while the precise linkages between cellular architecture and actomyosin motor organization remain unknown, our analysis suggests that the molecular basis of cell shape may, in addition to motor force, be a key adaptive strategy for malaria parasite dissemination and, as such, transmission.


Subject(s)
Biomechanical Phenomena , Plasmodium/cytology , Plasmodium/physiology , Actins/metabolism , Imaging, Three-Dimensional , Locomotion , Microscopy , Myosins/metabolism , Optical Imaging
19.
J Biol Chem ; 289(7): 4043-54, 2014 Feb 14.
Article in English | MEDLINE | ID: mdl-24371134

ABSTRACT

Actin depolymerizing factor (ADF)/cofilins are essential regulators of actin turnover in eukaryotic cells. These multifunctional proteins facilitate both stabilization and severing of filamentous (F)-actin in a concentration-dependent manner. At high concentrations ADF/cofilins bind stably to F-actin longitudinally between two adjacent actin protomers forming what is called a decorative interaction. Low densities of ADF/cofilins, in contrast, result in the optimal severing of the filament. To date, how these two contrasting modalities are achieved by the same protein remains uncertain. Here, we define the proximate amino acids between the actin filament and the malaria parasite ADF/cofilin, PfADF1 from Plasmodium falciparum. PfADF1 is unique among ADF/cofilins in being able to sever F-actin but do so without stable filament binding. Using chemical cross-linking and mass spectrometry (XL-MS) combined with structure reconstruction we describe a previously overlooked binding interface on the actin filament targeted by PfADF1. This site is distinct from the known binding site that defines decoration. Furthermore, total internal reflection fluorescence (TIRF) microscopy imaging of single actin filaments confirms that this novel low affinity site is required for F-actin severing. Exploring beyond malaria parasites, selective blocking of the decoration site with human cofilin (HsCOF1) using cytochalasin D increases its severing rate. HsCOF1 may therefore also use a decoration-independent site for filament severing. Thus our data suggest that a second, low affinity actin-binding site may be universally used by ADF/cofilins for actin filament severing.


Subject(s)
Destrin/chemistry , Plasmodium falciparum/chemistry , Protozoan Proteins/chemistry , Actin Cytoskeleton/chemistry , Actin Cytoskeleton/genetics , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/ultrastructure , Actins/chemistry , Actins/genetics , Actins/metabolism , Binding Sites , Cofilin 1/chemistry , Cofilin 1/genetics , Cofilin 1/metabolism , Cytochalasin D/chemistry , Destrin/genetics , Destrin/metabolism , Humans , Plasmodium falciparum/genetics , Plasmodium falciparum/metabolism , Protozoan Proteins/genetics , Protozoan Proteins/metabolism
20.
Cell ; 153(5): 1120-33, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23683579

ABSTRACT

Cell-cell communication is an important mechanism for information exchange promoting cell survival for the control of features such as population density and differentiation. We determined that Plasmodium falciparum-infected red blood cells directly communicate between parasites within a population using exosome-like vesicles that are capable of delivering genes. Importantly, communication via exosome-like vesicles promotes differentiation to sexual forms at a rate that suggests that signaling is involved. Furthermore, we have identified a P. falciparum protein, PfPTP2, that plays a key role in efficient communication. This study reveals a previously unidentified pathway of P. falciparum biology critical for survival in the host and transmission to mosquitoes. This identifies a pathway for the development of agents to block parasite transmission from the human host to the mosquito.


Subject(s)
Cell Communication , Erythrocytes/pathology , Erythrocytes/parasitology , Malaria, Falciparum/pathology , Malaria, Falciparum/parasitology , Plasmodium falciparum/physiology , Actins/antagonists & inhibitors , Animals , Culicidae/parasitology , Drug Resistance , Exosomes/parasitology , Humans , Microtubules/drug effects , Plasmids/genetics , Plasmodium falciparum/growth & development , Signal Transduction , Trophozoites/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...