Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 336
Filtrar
1.
mSphere ; 8(5): e0013123, 2023 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-37606582

RESUMO

Malaria parasites modify their host erythrocyte in multiple ways, leading to changes in the deformability, adhesiveness, and permeability of the host erythrocyte. Most of these changes are mediated by proteins exported from the parasite to the host erythrocyte, where these proteins interact with the host cell cytoskeleton or form complexes in the plasma membrane of the infected erythrocyte. In addition, malaria parasites induce the formation of membranous compartments-the parasitophorous vacuole, the tubovesicular network (TVN), the Maurer's clefts and small vesicles-within the infected erythrocyte, a cell that is normally devoid of internal membranes. After infection, changes also occur in the composition and asymmetry of the erythrocyte plasma membrane. Although many aspects of the mechanism of export of parasite proteins have become clear, the mechanism by which these membranous compartments are formed and expanded is almost entirely unknown. To determine whether parasite-derived phospholipids play a part in these processes, we applied a metabolic labeling technique that allows phosphatidylcholine to be labeled with a fluorophore. As the host erythrocyte cannot synthesize phospholipids, within infected erythrocytes, only parasite-derived phosphatidylcholine will be labeled with this technique. The results revealed that phosphatidylcholine produced by the parasite is distributed throughout the infected erythrocyte, including the TVN and the erythrocyte plasma membrane, but not Maurer's clefts. Interestingly, labeled phospholipids were also detected in the erythrocyte plasma membrane very soon after invasion of the parasites, indicating that the parasite may add phospholipids to the host erythrocyte during invasion. IMPORTANCE Here, we describe a previously unappreciated way in which the malaria parasite interacts with the host erythrocyte, namely, by the transfer of parasite phospholipids to the erythrocyte plasma membrane. This likely has important consequences for the survival of the parasite in the host cell and the host organism. We show that parasite-derived phospholipids are transferred from the parasite to the host erythrocyte plasma membrane and that other internal membranes that are produced after the parasite has invaded the cell are produced, at least in part, using parasite-derived phospholipids. The one exception to this is the Maurer's cleft, a membranous organelle that is involved in the transport of parasite proteins to the surface of the erythrocyte. This reveals that the Maurer's cleft is produced in a different manner than the other parasite-induced membranes. Overall, these findings provide a platform for the study of a new aspect of the host-parasite interaction.


Assuntos
Malária , Parasitos , Animais , Humanos , Fosfatidilcolinas/metabolismo , Plasmodium falciparum/metabolismo , Eritrócitos/parasitologia , Membrana Eritrocítica/metabolismo , Membrana Eritrocítica/parasitologia
2.
Sci Rep ; 12(1): 8934, 2022 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-35624125

RESUMO

Malaria parasites such as Plasmodium falciparum have exerted formidable selective pressures on the human genome. Of the human genetic variants associated with malaria protection, beta thalassaemia (a haemoglobinopathy) was the earliest to be associated with malaria prevalence. However, the malaria protective properties of beta thalassaemic erythrocytes remain unclear. Here we studied the mechanics and surface protein expression of beta thalassaemia heterozygous erythrocytes, measured their susceptibility to P. falciparum invasion, and calculated the energy required for merozoites to invade them. We found invasion-relevant differences in beta thalassaemic cells versus matched controls, specifically: elevated membrane tension, reduced bending modulus, and higher levels of expression of the major invasion receptor basigin. However, these differences acted in opposition to each other with respect to their likely impact on invasion, and overall we did not observe beta thalassaemic cells to have lower P. falciparum invasion efficiency for any of the strains tested.


Assuntos
Malária Falciparum , Malária , Talassemia beta , Membrana Eritrocítica/parasitologia , Heterozigoto , Humanos , Malária Falciparum/epidemiologia , Plasmodium falciparum/genética , Talassemia beta/genética
3.
Commun Biol ; 4(1): 1375, 2021 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-34880413

RESUMO

Cholesterol-rich microdomains are membrane compartments characterized by specific lipid and protein composition. These dynamic assemblies are involved in several biological processes, including infection by intracellular pathogens. This work provides a comprehensive analysis of the composition of human erythrocyte membrane microdomains. Based on their floating properties, we also categorized the microdomain-associated proteins into clusters. Interestingly, erythrocyte microdomains include the vast majority of the proteins known to be involved in invasion by the malaria parasite Plasmodium falciparum. We show here that the Ecto-ADP-ribosyltransferase 4 (ART4) and Aquaporin 1 (AQP1), found within one specific cluster, containing the essential host determinant CD55, are recruited to the site of parasite entry and then internalized to the newly formed parasitophorous vacuole membrane. By generating null erythroid cell lines, we showed that one of these proteins, ART4, plays a role in P. falciparum invasion. We also found that genetic variants in both ART4 and AQP1 are associated with susceptibility to the disease in a malaria-endemic population.


Assuntos
Membrana Eritrocítica/química , Eritrócitos/parasitologia , Malária Falciparum/parasitologia , Malária/parasitologia , Microdomínios da Membrana/química , Membrana Eritrocítica/parasitologia , Eritrócitos/química , Humanos , Plasmodium falciparum/fisiologia
4.
Proc Natl Acad Sci U S A ; 118(35)2021 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-34446549

RESUMO

The RhopH complex is implicated in malaria parasites' ability to invade and create new permeability pathways in host erythrocytes, but its mechanisms remain poorly understood. Here, we enrich the endogenous RhopH complex in a native soluble form, comprising RhopH2, CLAG3.1, and RhopH3, directly from parasite cell lysates and determine its atomic structure using cryo-electron microscopy (cryo-EM), mass spectrometry, and the cryoID program. CLAG3.1 is positioned between RhopH2 and RhopH3, which both share substantial binding interfaces with CLAG3.1 but make minimal contacts with each other. The forces stabilizing individual subunits include 13 intramolecular disulfide bonds. Notably, CLAG3.1 residues 1210 to 1223, previously predicted to constitute a transmembrane helix, are embedded within a helical bundle formed by residues 979 to 1289 near the C terminus of CLAG3.1. Buried in the core of the RhopH complex and largely shielded from solvent, insertion of this putative transmembrane helix into the erythrocyte membrane would likely require a large conformational rearrangement. Given the unusually high disulfide content of the complex, it is possible that such a rearrangement could be initiated by the breakage of allosteric disulfide bonds, potentially triggered by interactions at the erythrocyte membrane. This first direct observation of an exported Plasmodium falciparum transmembrane protein-in a soluble, trafficking state and with atomic details of buried putative membrane-insertion helices-offers insights into the assembly and trafficking of RhopH and other parasite-derived complexes to the erythrocyte membrane. Our study demonstrates the potential the endogenous structural proteomics approach holds for elucidating the molecular mechanisms of hard-to-isolate complexes in their native, functional forms.


Assuntos
Membrana Eritrocítica/metabolismo , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/química , Permeabilidade da Membrana Celular , Microscopia Crioeletrônica , Membrana Eritrocítica/parasitologia , Humanos , Modelos Moleculares , Nutrientes/metabolismo , Conformação Proteica , Proteômica , Proteínas de Protozoários/fisiologia , Proteínas de Protozoários/ultraestrutura , Relação Estrutura-Atividade
5.
J Exp Med ; 218(9)2021 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-34342640

RESUMO

We previously identified a Plasmodium falciparum (Pf) protein of unknown function encoded by a single-copy gene, PF3D7_1134300, as a target of antibodies in plasma of Tanzanian children in a whole-proteome differential screen. Here we characterize this protein as a blood-stage antigen that localizes to the surface membranes of both parasitized erythrocytes and merozoites, hence its designation as Pf erythrocyte membrane and merozoite antigen 1 (PfEMMA1). Mouse anti-PfEMMA1 antisera and affinity-purified human anti-PfEMMA1 antibodies inhibited growth of P. falciparum strains by up to 68% in growth inhibition assays. Following challenge with uniformly fatal Plasmodium berghei (Pb) ANKA, up to 40% of mice immunized with recombinant PbEMMA1 self-cured, and median survival of lethally infected mice was up to 2.6-fold longer than controls (21 vs. 8 d, P = 0.005). Furthermore, high levels of naturally acquired human anti-PfEMMA1 antibodies were associated with a 46% decrease in parasitemia over 2.5 yr of follow-up of Tanzanian children. Together, these findings suggest that antibodies to PfEMMA1 mediate protection against malaria.


Assuntos
Antígenos de Protozoários/metabolismo , Membrana Eritrocítica/parasitologia , Malária Falciparum/parasitologia , Merozoítos/metabolismo , Plasmodium falciparum/fisiologia , Proteínas de Protozoários/genética , Animais , Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/genética , Antígenos de Protozoários/imunologia , Pré-Escolar , Feminino , Interações Hospedeiro-Parasita/fisiologia , Humanos , Lactente , Vacinas Antimaláricas/genética , Vacinas Antimaláricas/imunologia , Malária Falciparum/imunologia , Malária Falciparum/mortalidade , Merozoítos/imunologia , Camundongos Endogâmicos BALB C , Plasmodium falciparum/imunologia , Plasmodium falciparum/patogenicidade , Polimorfismo de Nucleotídeo Único , Proteínas de Protozoários/química , Proteínas de Protozoários/imunologia , Proteínas de Protozoários/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Tanzânia
6.
Elife ; 102021 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-34286696

RESUMO

During the blood stage of malaria pathogenesis, parasites invade healthy red blood cells (RBC) to multiply inside the host and evade the immune response. When attached to RBC, the parasite first has to align its apex with the membrane for a successful invasion. Since the parasite's apex sits at the pointed end of an oval (egg-like) shape with a large local curvature, apical alignment is in general an energetically unfavorable process. Previously, using coarse-grained mesoscopic simulations, we have shown that optimal alignment time is achieved due to RBC membrane deformation and the stochastic nature of bond-based interactions between the parasite and RBC membrane (Hillringhaus et al., 2020). Here, we demonstrate that the parasite's shape has a prominent effect on the alignment process. The alignment times of spherical parasites for intermediate and large bond off-rates (or weak membrane-parasite interactions) are found to be close to those of an egg-like shape. However, for small bond off-rates (or strong adhesion and large membrane deformations), the alignment time for a spherical shape increases drastically. Parasite shapes with large aspect ratios such as oblate and long prolate ellipsoids are found to exhibit very long alignment times in comparison to the egg-like shape. At a stiffened RBC, a spherical parasite aligns faster than any other investigated shape. This study shows that the original egg-like shape performs not worse for parasite alignment than other considered shapes but is more robust with respect to different adhesion interactions and RBC membrane rigidities.


Assuntos
Adesão Celular/fisiologia , Membrana Eritrocítica/metabolismo , Eritrócitos/parasitologia , Hidrodinâmica , Plasmodium falciparum/fisiologia , Membrana Eritrocítica/parasitologia , Interações Hospedeiro-Parasita/fisiologia , Humanos , Malária Falciparum/parasitologia , Merozoítos/metabolismo
7.
Front Immunol ; 12: 643746, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34093532

RESUMO

Malaria remains one of the most serious health problems in developing countries. The causative agent of malaria, Plasmodium spp., have a complex life cycle involving multiple developmental stages as well as different morphological, biochemical and metabolic requirements. We recently found that γδ T cells control parasite growth using pore-forming proteins to deliver their cytotoxic proteases, the granzymes, into blood residing parasites. Here, we follow up on the molecular mechanisms of parasite growth inhibition by human pore-forming proteins. We confirm that Plasmodium falciparum infection efficiently depletes the red blood cells of cholesterol, which renders the parasite surrounding membranes susceptible to lysis by prokaryotic membrane disrupting proteins, such as lymphocytic granulysin or the human cathelicidin LL-37. Interestingly, not the cholesterol depletion but rather the simultaneous exposure of phosphatidylserine, a negatively charged phospholipid, triggers resistance of late stage parasitized red blood cells towards the eukaryotic pore forming protein perforin. Overall, by revealing the molecular events we establish here a pathogen-host interaction that involves host cell membrane remodeling that defines the susceptibility towards cytolytic molecules.


Assuntos
Membrana Eritrocítica/imunologia , Hemólise/imunologia , Malária Falciparum/imunologia , Perforina/imunologia , Plasmodium falciparum/imunologia , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Linfócitos T/imunologia , Antígenos de Diferenciação de Linfócitos T , Peptídeos Catiônicos Antimicrobianos/imunologia , Suscetibilidade a Doenças , Membrana Eritrocítica/parasitologia , Humanos , Catelicidinas
8.
Nat Commun ; 12(1): 3620, 2021 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-34131147

RESUMO

Host membrane remodeling is indispensable for viruses, bacteria, and parasites, to subvert the membrane barrier and obtain entry into cells. The malaria parasite Plasmodium spp. induces biophysical and molecular changes to the erythrocyte membrane through the ordered secretion of its apical organelles. To understand this process and address the debate regarding how the parasitophorous vacuole membrane (PVM) is formed, we developed an approach using lattice light-sheet microscopy, which enables the parasite interaction with the host cell membrane to be tracked and characterized during invasion. Our results show that the PVM is predominantly formed from the erythrocyte membrane, which undergoes biophysical changes as it is remodeled across all stages of invasion, from pre-invasion through to PVM sealing. This approach enables a functional interrogation of parasite-derived lipids and proteins in PVM biogenesis and echinocytosis during Plasmodium falciparum invasion and promises to yield mechanistic insights regarding how this is more generally orchestrated by other intracellular pathogens.


Assuntos
Membrana Eritrocítica/parasitologia , Eritrócitos/parasitologia , Tomografia Computadorizada Quadridimensional/métodos , Interações Hospedeiro-Parasita/fisiologia , Malária/parasitologia , Vacúolos/metabolismo , Animais , Membrana Eritrocítica/metabolismo , Humanos , Merozoítos , Parasitos , Plasmodium/metabolismo , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/metabolismo
9.
Nat Commun ; 12(1): 1792, 2021 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-33741926

RESUMO

In both sickle cell disease and malaria, red blood cells (RBCs) are phagocytosed in the spleen, but receptor-ligand pairs mediating uptake have not been identified. Here, we report that patches of high mannose N-glycans (Man5-9GlcNAc2), expressed on diseased or oxidized RBC surfaces, bind the mannose receptor (CD206) on phagocytes to mediate clearance. We find that extravascular hemolysis in sickle cell disease correlates with high mannose glycan levels on RBCs. Furthermore, Plasmodium falciparum-infected RBCs expose surface mannose N-glycans, which occur at significantly higher levels on infected RBCs from sickle cell trait subjects compared to those lacking hemoglobin S. The glycans are associated with high molecular weight complexes and protease-resistant, lower molecular weight fragments containing spectrin. Recognition of surface N-linked high mannose glycans as a response to cellular stress is a molecular mechanism common to both the pathogenesis of sickle cell disease and resistance to severe malaria in sickle cell trait.


Assuntos
Anemia Falciforme/metabolismo , Eritrócitos/metabolismo , Manose/metabolismo , Fagócitos/metabolismo , Polissacarídeos/metabolismo , Membrana Eritrocítica/metabolismo , Membrana Eritrocítica/parasitologia , Eritrócitos/parasitologia , Citometria de Fluxo/métodos , Hemólise , Humanos , Ligantes , Malária Falciparum/metabolismo , Malária Falciparum/parasitologia , Glicoproteínas de Membrana/metabolismo , Fagocitose , Plasmodium falciparum/fisiologia , Ligação Proteica , Receptores Imunológicos/metabolismo
10.
PLoS Pathog ; 17(2): e1009259, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33600495

RESUMO

The human malaria parasite Plasmodium falciparum relies on lipids to survive; this makes its lipid metabolism an attractive drug target. The lipid phosphatidylserine (PS) is usually confined to the inner leaflet of the red blood cell membrane (RBC) bilayer; however, some studies suggest that infection with the intracellular parasite results in the presence of this lipid in the RBC membrane outer leaflet, where it could act as a recognition signal to phagocytes. Here, we used fluorescent lipid analogues and probes to investigate the enzymatic reactions responsible for maintaining asymmetry between membrane leaflets, and found that in parasitised RBCs the maintenance of membrane asymmetry was partly disrupted, and PS was increased in the outer leaflet. We examined the underlying causes for the differences between uninfected and infected RBCs using fluorescent dyes and probes, and found that calcium levels increased in the infected RBC cytoplasm, whereas membrane cholesterol was depleted from the erythrocyte plasma membrane. We explored the resulting effect of PS exposure on enhanced phagocytosis by monocytes, and show that infected RBCs must expend energy to limit phagocyte recognition, and provide experimental evidence that PS exposure contributes to phagocytic recognition of P. falciparum-infected RBCs. Together, these findings underscore the pivotal role for PS exposure on the surface of Plasmodium falciparum-infected erythrocytes for in vivo interactions with the host immune system, and provide a rationale for targeted antimalarial drug design.


Assuntos
Cálcio/metabolismo , Membrana Eritrocítica/metabolismo , Eritrócitos/metabolismo , Malária Falciparum/metabolismo , Monócitos/metabolismo , Fagocitose , Fosfatidilserinas/metabolismo , Membrana Eritrocítica/parasitologia , Eritrócitos/parasitologia , Humanos , Malária Falciparum/parasitologia , Monócitos/parasitologia , Plasmodium falciparum/isolamento & purificação
11.
Exp Cell Res ; 397(2): 112370, 2020 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-33186602

RESUMO

The mechanical properties of erythrocytes have been investigated by different techniques. However, there are few reports on how the viscoelasticity of these cells varies during malaria disease. Here, we quantitatively map the viscoelastic properties of Plasmodium falciparum-parasitized human erythrocytes. We apply new methodologies based on optical tweezers to measure the viscoelastic properties and defocusing microscopy to measure the erythrocyte height profile, the overall cell volume, and its form factor, a crucial parameter to convert the complex elastic constant into complex shear modulus. The storage and loss shear moduli are obtained for each stage of parasite maturation inside red blood cells, while the former increase, the latter decrease. Employing a soft glassy rheology model, we obtain the power-law exponent for the storage and loss shear moduli, characterizing the soft glassy features of red blood cells in each parasite maturation stage. Ring forms present a liquid-like behavior, with a slightly lower power-law exponent than healthy erythrocytes, whereas trophozoite and schizont stages exhibit increasingly solid-like behaviors. Finally, the surface elastic shear moduli, low-frequency surface viscosities, and shape recovery relaxation times all increase not only in a stage-dependent manner but also when compared to healthy red blood cells. Overall, the results call attention to the soft glassy characteristics of Plasmodium falciparum-parasitized erythrocyte membrane and may provide a basis for future studies to better understand malaria disease from a mechanobiological perspective.


Assuntos
Módulo de Elasticidade , Membrana Eritrocítica/patologia , Eritrócitos Anormais/patologia , Eritrócitos/patologia , Malária/sangue , Plasmodium falciparum/crescimento & desenvolvimento , Viscosidade Sanguínea , Membrana Eritrocítica/parasitologia , Eritrócitos/parasitologia , Eritrócitos Anormais/parasitologia , Humanos , Malária/parasitologia , Plasmodium falciparum/patogenicidade , Reologia
12.
Parasitol Res ; 119(12): 4297-4302, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33089360

RESUMO

Malaria is caused by unicellular parasites of the genus Plasmodium, which reside in erythrocytes during the clinically relevant stage of infection. To separate parasite from host cell material, haemolytic agents such as saponin are widely used. Previous electron microscopy studies on saponin-treated parasites reported both, parasites enclosed by the erythrocyte membrane and liberated from the host cell. These ambiguous reports prompted us to investigate haemolysis by live-cell time-lapse microscopy. Using either saponin or streptolysin O to lyse Plasmodium falciparum-infected erythrocytes, we found that ring-stage parasites efficiently exit the erythrocyte upon haemolysis. For late-stage parasites, we found that only approximately half were freed, supporting the previous electron microscopy studies. Immunofluorescence imaging indicated that freed parasites were surrounded by the parasitophorous vacuolar membrane. These results may be of interest for future work using haemolytic agents to enrich for parasite material.


Assuntos
Eritrócitos/parasitologia , Hemólise/efeitos dos fármacos , Plasmodium falciparum/fisiologia , Saponinas/farmacologia , Estreptolisinas/farmacologia , Proteínas de Bactérias/farmacologia , Membrana Eritrocítica/efeitos dos fármacos , Membrana Eritrocítica/parasitologia , Eritrócitos/efeitos dos fármacos , Vesículas Extracelulares/parasitologia , Humanos , Estágios do Ciclo de Vida/fisiologia , Microscopia , Plasmodium falciparum/crescimento & desenvolvimento
13.
Malar J ; 19(1): 302, 2020 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-32847585

RESUMO

BACKGROUND: The intra-erythrocytic development of the malaria parasite Plasmodium falciparum depends on the uptake of a number of essential nutrients from the host cell and blood plasma. It is widely recognized that the parasite imports low molecular weight solutes from the plasma and the consumption of these nutrients by P. falciparum has been extensively analysed. However, although it was already shown that the parasite also imports functional proteins from the vertebrate host, the internalization route through the different infected erythrocyte membranes has not yet been elucidated. In order to further understand the uptake mechanism, the study examined the trafficking of human plasminogen from the extracellular medium into P. falciparum-infected red blood cells. METHODS: Plasmodium falciparum clone 3D7 was cultured in standard HEPES-buffered RPMI 1640 medium supplemented with 0.5% AlbuMAX. Exogenous human plasminogen was added to the P. falciparum culture and the uptake of this protein by the parasites was analysed by electron microscopy and Western blotting. Immunoprecipitation and mass spectrometry were performed to investigate possible protein interactions that may assist plasminogen import into infected erythrocytes. The effect of pharmacological inhibitors of different cellular physiological processes in plasminogen uptake was also tested. RESULTS: It was observed that plasminogen was selectively internalized by P. falciparum-infected erythrocytes, with localization in plasma membrane erythrocyte and parasite's cytosol. The protein was not detected in parasitic food vacuole and haemoglobin-containing vesicles. Furthermore, in erythrocyte cytoplasm, plasminogen was associated with the parasite-derived membranous structures tubovesicular network (TVN) and Maurer's clefts. Several proteins were identified in immunoprecipitation assay and may be involved in the delivery of plasminogen across the P. falciparum multiple compartments. CONCLUSION: The findings here reported reveal new features regarding the acquisition of plasma proteins of the host by P. falciparum-infected erythrocytes, a mechanism that involves the exomembrane system, which is distinct from the haemoglobin uptake, clarifying a route that may be potentially targeted for inhibition studies.


Assuntos
Eritrócitos/parasitologia , Plasminogênio/metabolismo , Plasmodium falciparum/fisiologia , Membrana Eritrocítica/parasitologia , Interações Hospedeiro-Parasita , Humanos , Malária Falciparum/parasitologia , Plasma/química , Transporte Proteico
14.
Elife ; 92020 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-32420874

RESUMO

Malaria parasites invade healthy red blood cells (RBCs) during the blood stage of the disease. Even though parasites initially adhere to RBCs with a random orientation, they need to align their apex toward the membrane in order to start the invasion process. Using hydrodynamic simulations of a RBC and parasite, where both interact through discrete stochastic bonds, we show that parasite alignment is governed by the combination of RBC membrane deformability and dynamics of adhesion bonds. The stochastic nature of bond-based interactions facilitates a diffusive-like re-orientation of the parasite at the RBC membrane, while RBC deformation aids in the establishment of apex-membrane contact through partial parasite wrapping by the membrane. This bond-based model for parasite adhesion quantitatively captures alignment times measured experimentally and demonstrates that alignment times increase drastically with increasing rigidity of the RBC membrane. Our results suggest that the alignment process is mediated simply by passive parasite adhesion.


Assuntos
Adesão Celular/fisiologia , Membrana Eritrocítica/metabolismo , Eritrócitos/parasitologia , Hidrodinâmica , Plasmodium falciparum/fisiologia , Membrana Eritrocítica/parasitologia , Interações Hospedeiro-Parasita/fisiologia , Humanos , Malária Falciparum/parasitologia , Merozoítos/metabolismo
15.
mBio ; 11(2)2020 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-32184257

RESUMO

The malaria parasite Plasmodium falciparum traffics the virulence protein P. falciparum erythrocyte membrane protein 1 (PfEMP1) to the surface of infected red blood cells (RBCs) via membranous organelles, known as the Maurer's clefts. We developed a method for efficient enrichment of Maurer's clefts and profiled the protein composition of this trafficking organelle. We identified 13 previously uncharacterized or poorly characterized Maurer's cleft proteins. We generated transfectants expressing green fluorescent protein (GFP) fusions of 7 proteins and confirmed their Maurer's cleft location. Using co-immunoprecipitation and mass spectrometry, we generated an interaction map of proteins at the Maurer's clefts. We identified two key clusters that may function in the loading and unloading of PfEMP1 into and out of the Maurer's clefts. We focus on a putative PfEMP1 loading complex that includes the protein GEXP07/CX3CL1-binding protein 2 (CBP2). Disruption of GEXP07 causes Maurer's cleft fragmentation, aberrant knobs, ablation of PfEMP1 surface expression, and loss of the PfEMP1-mediated adhesion. ΔGEXP07 parasites have a growth advantage compared to wild-type parasites, and the infected RBCs are more deformable and more osmotically fragile.IMPORTANCE The trafficking of the virulence antigen PfEMP1 and its presentation at the knob structures at the surface of parasite-infected RBCs are central to severe adhesion-related pathologies such as cerebral and placental malaria. This work adds to our understanding of how PfEMP1 is trafficked to the RBC membrane by defining the protein-protein interaction networks that function at the Maurer's clefts controlling PfEMP1 loading and unloading. We characterize a protein needed for virulence protein trafficking and provide new insights into the mechanisms for host cell remodeling, parasite survival within the host, and virulence.


Assuntos
Membrana Eritrocítica/metabolismo , Eritrócitos/citologia , Interações Hospedeiro-Parasita , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/metabolismo , Proteínas de Transporte/metabolismo , Linhagem Celular , Membrana Eritrocítica/parasitologia , Eritrócitos/parasitologia , Humanos , Proteínas de Membrana , Plasmodium falciparum/genética , Plasmodium falciparum/patogenicidade , Mapas de Interação de Proteínas , Transporte Proteico , Proteínas de Protozoários/genética
16.
Georgian Med News ; (294): 103-108, 2019 Sep.
Artigo em Russo | MEDLINE | ID: mdl-31687959

RESUMO

The article overviews some issues of the severe course of tropical malaria. In addition to the analysis of the ongoing situation with malaria in Russia, a general clinical picture of the severe course of tropical malaria is discussed. The main part of the overview includes a detailed analysis of current data on the molecular genetic aspects of the erythrocytes' adhesion in the case of tropical malaria. The main elements involved in the process of binding red blood cells and, as a result, in the process of their adhesion to other cells of the human body were considered in detail. Data were studied and summarized not only on protein interactions between an infected red cell and its cellular environment, but also on the genetic characteristics of the parasite leading to similar molecular-biological processes. In addition to the study of protein PfEMP1 role which is nowadays well-considered in the literature, the most up-to-date but less reported data on erythrocyte adhesion proteins STEVOR and RIFIN were also included. The team of authors hopes that this publication will help to get a deeper insight into the problem of erythrocyte adhesion in the course of complicated malaria infection forms and to summarize some of the available data on this issue.


Assuntos
Antígenos de Protozoários/metabolismo , Membrana Eritrocítica/química , Membrana Eritrocítica/parasitologia , Eritrócitos/parasitologia , Malária Falciparum , Proteínas de Membrana/metabolismo , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/metabolismo , Antígenos de Protozoários/química , Humanos , Malária Falciparum/sangue , Proteínas de Membrana/química , Plasmodium falciparum/fisiologia , Proteínas de Protozoários/química , Federação Russa
17.
Commun Biol ; 2: 311, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31428699

RESUMO

During intraerythrocytic development, the human malaria parasite Plasmodium falciparum alters the mechanical deformability of its host cell. The underpinning biological processes involve gain in parasite mass, changes in the membrane protein compositions, reorganization of the cytoskeletons and its coupling to the plasma membrane, and formation of membrane protrusions, termed knobs. The hemoglobinopathies S and C are known to partially protect carriers from severe malaria, possibly through additional changes in the erythrocyte biomechanics, but a detailed quantification of cell mechanics is still missing. Here, we combined flicker spectroscopy and a mathematical model and demonstrated that knob formation strongly suppresses membrane fluctuations by increasing membrane-cytoskeleton coupling. We found that the confinement increased with hemoglobin S but decreases with hemoglobin C in spite of comparable knob densities and diameters. We further found that the membrane bending modulus strongly depends on the hemoglobinopathetic variant, suggesting increased amounts of irreversibly oxidized hemichromes bound to membranes.


Assuntos
Membrana Eritrocítica/parasitologia , Hemoglobina C/metabolismo , Hemoglobina Falciforme/metabolismo , Plasmodium falciparum/fisiologia , Fenômenos Biomecânicos , Simulação por Computador , Humanos , Mutação/genética , Análise Numérica Assistida por Computador
18.
PLoS Pathog ; 15(7): e1007870, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31260501

RESUMO

Naturally acquired clinical immunity to Plasmodium falciparum is partly mediated by antibodies directed at parasite-derived antigens expressed on the surface of red blood cells which mediate disease and are extremely diverse. Unlike children, adults recognize a broad range of variant surface antigens (VSAs) and are protected from severe disease. Though crucial to the design and feasibility of an effective malaria vaccine, it is not yet known whether immunity arises through cumulative exposure to each of many antigenic types, cross-reactivity between antigenic types, or some other mechanism. In this study, we measured plasma antibody responses of 36 children with symptomatic malaria to a diverse panel of 36 recombinant proteins comprising part of the DBLα domain (the 'DBLα-tag') of PfEMP1, a major class of VSAs. We found that although plasma antibody responses were highly specific to individual antigens, serological profiles of responses across antigens fell into one of just two distinct types. One type was found almost exclusively in children that succumbed to severe disease (19 out of 20) while the other occurred in all children with mild disease (16 out of 16). Moreover, children with severe malaria had serological profiles that were narrower in antigen specificity and shorter-lived than those in children with mild malaria. Borrowing a novel technique used in influenza-antigenic cartography-we mapped these dichotomous serological profiles to amino acid sequence variation within a small sub-region of the PfEMP1 DBLα domain. By applying our methodology on a larger scale, it should be possible to identify epitopes responsible for eliciting the protective version of serological profiles to PfEMP1 thereby accelerating development of a broadly effective anti-disease malaria vaccine.


Assuntos
Antígenos de Protozoários/imunologia , Plasmodium falciparum/imunologia , Plasmodium falciparum/patogenicidade , Proteínas de Protozoários/imunologia , Sequência de Aminoácidos , Anticorpos Antiprotozoários/sangue , Variação Antigênica , Antígenos de Protozoários/genética , Pré-Escolar , Epitopos/genética , Epitopos/imunologia , Membrana Eritrocítica/imunologia , Membrana Eritrocítica/parasitologia , Feminino , Humanos , Lactente , Malária Falciparum/imunologia , Malária Falciparum/parasitologia , Masculino , Plasmodium falciparum/genética , Proteínas de Protozoários/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Alinhamento de Sequência
19.
PLoS Pathog ; 15(5): e1007761, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31071194

RESUMO

Plasmodium falciparum mediates adhesion of infected red blood cells (RBCs) to blood vessel walls by assembling a multi-protein complex at the RBC surface. This virulence-mediating structure, called the knob, acts as a scaffold for the presentation of the major virulence antigen, P. falciparum Erythrocyte Membrane Protein-1 (PfEMP1). In this work we developed correlative STochastic Optical Reconstruction Microscopy-Scanning Electron Microscopy (STORM-SEM) to spatially and temporally map the delivery of the knob-associated histidine-rich protein (KAHRP) and PfEMP1 to the RBC membrane skeleton. We show that KAHRP is delivered as individual modules that assemble in situ, giving a ring-shaped fluorescence profile around a dimpled disk that can be visualized by SEM. Electron tomography of negatively-stained membranes reveals a previously observed spiral scaffold underpinning the assembled knobs. Truncation of the C-terminal region of KAHRP leads to loss of the ring structures, disruption of the raised disks and aberrant formation of the spiral scaffold, pointing to a critical role for KAHRP in assembling the physical knob structure. We show that host cell actin remodeling plays an important role in assembly of the virulence complex, with cytochalasin D blocking knob assembly. Additionally, PfEMP1 appears to be delivered to the RBC membrane, then inserted laterally into knob structures.


Assuntos
Membrana Eritrocítica/parasitologia , Eritrócitos/parasitologia , Malária Falciparum/parasitologia , Peptídeos/metabolismo , Plasmodium falciparum/patogenicidade , Proteínas de Protozoários/metabolismo , Membrana Eritrocítica/metabolismo , Eritrócitos/metabolismo , Humanos , Malária Falciparum/metabolismo , Microscopia Eletrônica de Varredura , Peptídeos/química , Proteínas de Protozoários/química , Virulência
20.
Cell Microbiol ; 21(5): e12999, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30597708

RESUMO

Ferlins mediate calcium-dependent vesicular fusion. Although conserved throughout eukaryotic evolution, their function in unicellular organisms including apicomplexan parasites is largely unknown. Here, we define a crucial role for a ferlin-like protein (FLP) in host-to-vector transmission of the rodent malaria parasite Plasmodium berghei. Infection of the mosquito vectors requires the formation of free gametes and their fertilisation in the mosquito midgut. Mature gametes will only emerge upon secretion of factors that stimulate the disruption of the red blood cell membrane and the parasitophorous vacuole membrane. Genetic depletion of FLP in sexual stages leads to a complete life cycle arrest in the mosquito. Although mature gametes form normally, mutants lacking FLP remain trapped in the red blood cell. The egress defect is rescued by detergent-mediated membrane lysis. In agreement with ferlin vesicular localisation, HA-tagged FLP labels intracellular speckles, which relocalise to the cell periphery during gamete maturation. Our data define FLP as a novel critical factor for Plasmodium fertilisation and transmission and suggest an evolutionarily conserved example of ferlin-mediated exocytosis.


Assuntos
Membrana Eritrocítica/metabolismo , Eritrócitos/metabolismo , Células Germinativas/metabolismo , Malária/transmissão , Plasmodium berghei/crescimento & desenvolvimento , Proteínas de Protozoários/metabolismo , Animais , Culicidae/parasitologia , Detergentes/farmacologia , Membrana Eritrocítica/efeitos dos fármacos , Membrana Eritrocítica/genética , Membrana Eritrocítica/parasitologia , Eritrócitos/efeitos dos fármacos , Eritrócitos/parasitologia , Exocitose/genética , Feminino , Células Germinativas/citologia , Células Germinativas/crescimento & desenvolvimento , Células Germinativas/ultraestrutura , Interações Hospedeiro-Patógeno , Estágios do Ciclo de Vida/genética , Malária/genética , Malária/metabolismo , Malária/parasitologia , Camundongos , Camundongos Endogâmicos C57BL , Mosquitos Vetores/genética , Mosquitos Vetores/metabolismo , Plasmodium berghei/genética , Plasmodium berghei/patogenicidade , Domínios Proteicos/genética , Proteínas de Protozoários/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...