Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 143
Filter
1.
Acta Trop ; 207: 105464, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32302689

ABSTRACT

Toxoplasma gondii is an obligatory intracellular parasite that critically depends on active invasion and egress from infected host cells to complete its propagation cycle. T. gondii rhoptry proteins (TgROPs) are virulent factors associated with host cell invasion, growth. In this study, we analyzed the functions of ROP9 in the process of T. gondii infection. The TgROP9 knockout RH strain (RH△ROP9) and its recovery strain (RH-ReROP9) were constructed using the CRISPR/Cas9 system. The invasion, proliferation, and egress efficiency of the RH△ROP9 strain were evaluated and their pathogenicity to mice was analyzed. Compared with RH wild-type (RH-WT) and RH-ReROP9 strains, the invasion percentage of RH△ROP9 to Vero cells was reduced by about 28.0% (p< 0.01) at 1.5 h, and the relative proliferation percentage was decreased by about 35.0% (p< 0.01) after infection with 102 or 103 parasites. In addition, the RH△ROP9 strain also showed prolonged egress time from host cells. The survival time of the mice (12.6 ± 1.6 or 10.1 ± 1.1 days) were delayed (p < 0.001) after infection with either 200 or 1000 RH△ROP9 parasites. These evidences suggested that ROP9 facilitated T. gondii infection in vitro and in vivo.


Subject(s)
Antigens, Protozoan/physiology , Membrane Proteins/physiology , Toxoplasma/pathogenicity , Animals , Chlorocebus aethiops , Female , Mice , Mice, Inbred BALB C , Vero Cells
2.
J Microbiol Biotechnol ; 27(10): 1844-1854, 2017 Oct 28.
Article in English | MEDLINE | ID: mdl-28838225

ABSTRACT

Trichomonas vaginalis is a pathogen that triggers severe immune responses in hosts. T. vaginalis α-actinin 2 (Tvα-actinin 2) has been used to diagnose trichomoniasis. Tvα-actinin 2 was dissected into three parts; the N-terminal, central, and C-terminal portions of the protein (#1, #2, and #3, respectively). Western blot of these Tvα-actinin 2 proteins with pooled patients' sera indicated that #2 and #3, but not #1, reacted with those sera. Immunofluorescence assays of two different forms of T. vaginalis (trophozoites and amoeboid forms), using anti-Tvα-actinin 2 antibodies, showed localization of Tvα-actinin 2 close to the plasma membranes of the amoeboid form. Fractionation experiments indicated the presence of Tvα-actinin 2 in cytoplasmic, membrane, and secreted proteins of T. vaginalis. Binding of fluorescence-labeled Trichomonas to vaginal epithelial cells and prostate cells was decreased in the antibody blocking experiment using anti-Tvα-actinin 2 antibodies. Pretreatment of T. vaginalis with anti-rTvα-actinin 2 antibodies also resulted in reduction in its cytotoxicity. Flow cytometry, ligand-binding immunoblotting assay, and observation by fluorescence microscopy were used to detect the binding of recombinant Tvα-actinin 2 to human epithelial cell lines. Specifically, the truncated N-terminal portion of Tvα-actinin 2, Tvα-actinin 2 #1, was shown to bind directly to vaginal epithelial cells. These data suggest that α-actinin 2 is one of the virulence factors responsible for the pathogenesis of T. vaginalis by serving as an adhesin to the host cells.


Subject(s)
Actinin/physiology , Trichomonas vaginalis/metabolism , Actinin/genetics , Antigens, Protozoan/genetics , Antigens, Protozoan/physiology , Carrier Proteins/genetics , Carrier Proteins/physiology , Cell Line , Epithelial Cells , Female , Gene Expression Regulation , Humans , Recombinant Proteins , Trichomonas Infections/immunology , Trichomonas vaginalis/genetics , Trichomonas vaginalis/immunology , Trophozoites , Vagina , Virulence Factors
3.
Parasit Vectors ; 10(1): 251, 2017 May 19.
Article in English | MEDLINE | ID: mdl-28526096

ABSTRACT

BACKGROUND: Adhesin proteins are used by Plasmodium parasites to bind and invade target cells. Hence, characterising molecules that participate in reticulocyte interaction is key to understanding the molecular basis of Plasmodium vivax invasion. This study focused on predicting functionally restricted regions of the P. vivax GPI-anchored micronemal antigen (PvGAMA) and characterising their reticulocyte binding activity. RESULTS: The pvgama gene was initially found in P. vivax VCG-I strain schizonts. According to the genetic diversity analysis, PvGAMA displayed a size polymorphism very common for antigenic P. vivax proteins. Two regions along the antigen sequence were highly conserved among species, having a negative natural selection signal. Interestingly, these regions revealed a functional role regarding preferential target cell adhesion. CONCLUSIONS: To our knowledge, this study describes PvGAMA reticulocyte binding properties for the first time. Conserved functional regions were predicted according to natural selection analysis and their binding ability was confirmed. These findings support the notion that PvGAMA may have an important role in P. vivax merozoite adhesion to its target cells.


Subject(s)
Conserved Sequence/physiology , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Reticulocytes/parasitology , Selection, Genetic , Antigens, Protozoan/genetics , Antigens, Protozoan/physiology , Cell Adhesion , Genetic Variation , Plasmodium vivax/genetics , Polymorphism, Genetic , Protein Binding , Sequence Analysis, DNA
4.
Trends Parasitol ; 31(7): 285-6, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25959958

ABSTRACT

The binding of multiple uninfected erythrocytes to a central malaria parasite-infected erythrocyte (IE) is called rosetting. Rosetting has been associated with severe disease, but its functional significance,and the host receptors and parasite ligands involved are only partially known. A recent study, which describes yet another piece in this already complex puzzle, provides a welcome boost and a broadening of an important malaria research field.


Subject(s)
Antigens, Protozoan/physiology , Erythrocytes/parasitology , Malaria, Falciparum/metabolism , Plasmodium falciparum/metabolism , Protozoan Proteins/physiology , Animals , Humans , Male
6.
Nat Med ; 21(4): 314-7, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25751816

ABSTRACT

Rosetting is a virulent Plasmodium falciparum phenomenon associated with severe malaria. Here we demonstrate that P. falciparum-encoded repetitive interspersed families of polypeptides (RIFINs) are expressed on the surface of infected red blood cells (iRBCs), where they bind to RBCs--preferentially of blood group A--to form large rosettes and mediate microvascular binding of iRBCs. We suggest that RIFINs have a fundamental role in the development of severe malaria and thereby contribute to the varying global distribution of ABO blood groups in the human population.


Subject(s)
Antigens, Protozoan/physiology , Erythrocytes/parasitology , Malaria, Falciparum/metabolism , Plasmodium falciparum/metabolism , Protozoan Proteins/physiology , ABO Blood-Group System , Animals , CHO Cells , Cricetinae , Cricetulus , Dogs , Drosophila , Escherichia coli/metabolism , Fluorescent Antibody Technique, Indirect , Humans , Immunoglobulin G/immunology , Male , Microcirculation , Microscopy, Confocal , Microsomes/metabolism , Pancreas/parasitology , Protein Multimerization , Rats , Rats, Sprague-Dawley , Sequence Analysis, RNA , Transfection
7.
Infect Immun ; 82(11): 4842-53, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25156731

ABSTRACT

Clinical immunity to pregnancy associated-malaria (PAM) in multigravida women has been attributed to antibodies that recognize VAR2CSA on the infected erythrocyte (IE) surface. The size and complexity of VAR2CSA have focused efforts on selecting one or more of its six Duffy binding-like (DBL) domains for vaccine development. Presently, however, there is no consensus as to which DBL domain(s) would be most effective in eliciting immunity. This is because antibodies to a number of the DBL domains have been found to block the adhesion of VAR2CSA-expressing erythrocytes to chondroitin sulfate A (CSA)-a major criterion for evaluating vaccine candidacy. Opsonization of IEs by cytophilic antibodies that recognize VAR2CSA represents an important yet understudied effector mechanism in acquired immunity to PAM. To date, no studies have sought to determine the targets of those antibodies. In this study, we found that IgGs from multigravida Malian women showed (i) higher reactivity to recombinant DBL domains by enzyme-linked immunosorbent assay (ELISA), (ii) more binding to VAR2CSA-expressing IEs, and (iii) greater opsonization of these IEs by human monocytic cells than IgGs from malaria-exposed Malian men and malaria-naive American adults. Preincubation of IgGs from multigravida women with recombinant DBL2χ, DBL3χ, or DBL5ε domains significantly diminished opsonization of VAR2CSA-expressing IEs by human monocytes. These data identify the DBL2χ, DBL3χ, and DBL5ε domains as the primary targets of opsonizing IgGs for the first time. Our study introduces a new approach to determining the antigenic targets of opsonizing IgGs in phagocytosis assays.


Subject(s)
Antibodies, Protozoan/immunology , Antigens, Protozoan/physiology , Immunoglobulin G/immunology , Malaria/immunology , Opsonin Proteins/metabolism , Pregnancy Complications, Parasitic/immunology , Antibody Affinity , Female , Humans , Malaria/blood , Male , Mali/epidemiology , Pregnancy , Receptors, IgG , United States
8.
Infect Immun ; 82(5): 1860-71, 2014 May.
Article in English | MEDLINE | ID: mdl-24566620

ABSTRACT

Protective immunity to Plasmodium falciparum malaria acquired after natural exposure is largely antibody mediated. IgG-specific P. falciparum EMP1 (PfEMP1) proteins on the infected erythrocyte surface are particularly important. The transient antibody responses and the slowly acquired protective immunity probably reflect the clonal antigenic variation and allelic polymorphism of PfEMP1. However, it is likely that other immune-evasive mechanisms are also involved, such as interference with formation and maintenance of immunological memory. We measured PfEMP1-specific antibody levels by enzyme-linked immunosorbent assay (ELISA) and memory B-cell frequencies by enzyme-linked immunosorbent spot (ELISPOT) assay in a cohort of P. falciparum-exposed nonpregnant Ghanaian women. The antigens used were a VAR2CSA-type PfEMP1 (IT4VAR04) with expression restricted to parasites infecting the placenta, as well as two commonly recognized PfEMP1 proteins (HB3VAR06 and IT4VAR60) implicated in rosetting and not pregnancy restricted. This enabled, for the first time, a direct comparison in the same individuals of immune responses specific for a clinically important parasite antigen expressed only during well-defined periods (pregnancy) to responses specific for comparable antigens expressed independent of pregnancy. Our data indicate that PfEMP1-specific B-cell memory is adequately acquired even when antigen exposure is infrequent (e.g., VAR2CSA-type PfEMP1). Furthermore, immunological memory specific for VAR2CSA-type PfEMP1 can be maintained for many years without antigen reexposure and after circulating antigen-specific IgG has disappeared. The study provides evidence that natural exposure to P. falciparum leads to formation of durable B-cell immunity to clinically important PfEMP1 antigens. This has encouraging implications for current efforts to develop PfEMP1-based vaccines.


Subject(s)
Antigens, Protozoan/physiology , B-Lymphocytes/physiology , Malaria, Falciparum/immunology , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Adult , Antibodies, Protozoan/physiology , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression Regulation/physiology , Ghana/epidemiology , Humans , Immunoglobulin G/blood , Malaria, Falciparum/epidemiology , Pregnancy , Pregnancy Complications, Parasitic/epidemiology , Pregnancy Complications, Parasitic/immunology , Protozoan Proteins/genetics
9.
Infect Immun ; 81(10): 3825-34, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23897612

ABSTRACT

Following Anopheles mosquito-mediated introduction into a human host, Plasmodium parasites infect hepatocytes and undergo intensive replication. Accumulating evidence indicates that CD8(+) T cells induced by immunization with attenuated Plasmodium sporozoites can confer sterile immunity at the liver stage of infection; however, the mechanisms underlying this protection are not clearly understood. To address this, we generated recombinant Plasmodium berghei ANKA expressing a fusion protein of an ovalbumin epitope and green fluorescent protein in the cytoplasm of the parasite. We have shown that the ovalbumin epitope is presented by infected liver cells in a manner dependent on a transporter associated with antigen processing and becomes a target of specific CD8(+) T cells from the T cell receptor transgenic mouse line OT-I, leading to protection at the liver stage of Plasmodium infection. We visualized the interaction between OT-I cells and infected hepatocytes by intravital imaging using two-photon microscopy. OT-I cells formed clusters around infected hepatocytes, leading to the elimination of the intrahepatic parasites and subsequent formation of large clusters of OT-I cells in the liver. Gamma interferon expressed in CD8(+) T cells was dispensable for this protective response. Additionally, we found that polyclonal ovalbumin-specific memory CD8(+) T cells induced by de novo immunization were able to confer sterile protection, although the threshold frequency of the protection was relatively high. These studies revealed a novel mechanism of specific CD8(+) T cell-mediated protective immunity and demonstrated that proteins expressed in the cytoplasm of Plasmodium parasites can become targets of specific CD8(+) T cells during liver-stage infection.


Subject(s)
Antigens, Protozoan/physiology , CD8-Positive T-Lymphocytes/physiology , Hepatocytes/parasitology , Plasmodium berghei/metabolism , Animals , Gene Expression Regulation , Humans , Liver , Malaria , Mice , Mice, Transgenic , Nucleoproteins , Plasmodium berghei/genetics , Protozoan Proteins/genetics , Protozoan Proteins/metabolism
10.
Exp Parasitol ; 132(2): 109-15, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22691540

ABSTRACT

Leishmania is an obligate intracellular protozoan parasite that infects cells of the reticulo-endothelial system. Host defences against Leishmania include fever and oxidant production, and the parasite has developed a number of defence mechanisms to neutralize the host response. The Leishmania donovani A2 family of proteins has been shown to be essential for survival in mammalian visceral organs. Here we provide evidence that A2 proteins protect the parasite against host defences, namely heat stress (fever) and oxidative stress. A2 is however unable to protect the cells from endoplasmic reticulum stress induced by dithiothreitol. To downregulate A2 protein expression, L. donovani was transfected with an A2 antisense RNA expressing-vector, resulting in significant reduction of A2 levels. The resulting A2-deficient cells were more sensitive to heat shock and this was associated with increased production of internal oxidants during heat shock. Moreover, axenic amastigotes with downregulated A2 expression had increased internal oxidants and decreased viability following treatment with hydrogen peroxide or a nitric oxide donor when compared to control cells. Overall, these results suggest that A2 protects L. donovani from a variety of stresses, thereby allowing it to survive in the internal organs of the mammalian host and to cause visceral disease.


Subject(s)
Antigens, Protozoan/physiology , Heat-Shock Response/physiology , Leishmania donovani/pathogenicity , Oxidative Stress/physiology , Protozoan Proteins/physiology , Virulence Factors/physiology , Antigens, Protozoan/analysis , Axenic Culture , Down-Regulation , Leishmania donovani/chemistry , Plasmids , Protozoan Proteins/analysis , Tubulin/analysis , Virulence Factors/analysis
12.
PLoS Pathog ; 7(2): e1001282, 2011 Feb 10.
Article in English | MEDLINE | ID: mdl-21347354

ABSTRACT

Host cell invasion by apicomplexan parasites requires formation of the moving junction (MJ), a ring-like apposition between the parasite and host plasma membranes that the parasite migrates through during entry. The Toxoplasma MJ is a secreted complex including TgAMA1, a transmembrane protein on the parasite surface, and a complex of rhoptry neck proteins (TgRON2/4/5/8) described as host cell-associated. How these proteins connect the parasite and host cell has not previously been described. Here we show that TgRON2 localizes to the MJ and that two short segments flanking a hydrophobic stretch near its C-terminus (D3 and D4) independently associate with the ectodomain of TgAMA1. Pre-incubation of parasites with D3 (fused to glutathione S-transferase) dramatically reduces invasion but does not prevent injection of rhoptry bulb proteins. Hence, the entire C-terminal region of TgRON2 forms the crucial bridge between TgAMA1 and the rest of the MJ complex but this association is not required for rhoptry protein injection.


Subject(s)
Antigens, Protozoan/metabolism , Host-Parasite Interactions , Protozoan Proteins/chemistry , Protozoan Proteins/physiology , Toxoplasma/physiology , Amino Acid Motifs , Animals , Antigens, Protozoan/physiology , Cells, Cultured , Host-Parasite Interactions/immunology , Host-Parasite Interactions/physiology , Humans , Protein Binding/physiology , Protein Structure, Tertiary/physiology , Protozoan Proteins/immunology , Protozoan Proteins/metabolism , Toxoplasma/immunology , Toxoplasma/metabolism , Toxoplasmosis/parasitology , Toxoplasmosis/pathology
13.
Proc Natl Acad Sci U S A ; 107(38): 16643-8, 2010 Sep 21.
Article in English | MEDLINE | ID: mdl-20823248

ABSTRACT

Plasmodium falciparum erythrocyte membrane protein 1 (PfEMP1) family members mediate the adherence of parasite-infected red blood cells (IRBCs) to various host receptors. A previous study has shown that the parasite protein, cytoadherence-linked asexual gene 9 (CLAG9), is also essential for IRBC adherence. However, how CLAG9 influences this process remains unknown. In this study, we show that CLAG9 interacts with VAR2CSA, a PfEMP1 that mediates IRBC adherence to chondroitin 4-sulfate in the placenta. Importantly, our results show that the adherent parasites synthesize CLAG9 at two stages--the early ring and late trophozoite stages. Localization studies revealed that a substantial level of CLAG9 is located mainly at or in close proximity of the IRBC membrane in association with VAR2CSA. Upon treatment of IRBCs with trypsin, a significant amount of CLAG9 (≈150 kDa) was converted into ≈142-kDa polypeptide. Together these data demonstrate that a considerable amount of CLAG9 is embedded in the IRBC membrane such that at least a portion of the polypeptide at either N or C terminus is exposed on the cell surface. In parasites lacking CLAG9, VAR2CSA failed to express on the IRBC surface and was located within the parasite. Based on these findings, we propose that CLAG9 plays a critical role in the trafficking of PfEMP1s onto the IRBC surface. These results have important implications for the development of therapeutics for cerebral, placental, and other cytoadherence-associated malaria illnesses.


Subject(s)
Antigens, Protozoan/physiology , Cell Adhesion Molecules/physiology , Plasmodium falciparum/physiology , Plasmodium falciparum/pathogenicity , Protozoan Proteins/physiology , Animals , Antigens, Protozoan/chemistry , Antigens, Protozoan/genetics , Base Sequence , Cell Adhesion/physiology , Cell Adhesion Molecules/biosynthesis , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/genetics , Chondroitin Sulfates/physiology , DNA, Protozoan/genetics , Erythrocyte Membrane/parasitology , Erythrocyte Membrane/physiology , Erythrocyte Membrane/ultrastructure , Erythrocytes/parasitology , Female , Gene Knockout Techniques , Genes, Protozoan , Host-Parasite Interactions/physiology , Humans , In Vitro Techniques , Microscopy, Immunoelectron , Multiprotein Complexes , Placenta/parasitology , Placenta/physiology , Plasmodium falciparum/genetics , Plasmodium falciparum/growth & development , Pregnancy , Protozoan Proteins/biosynthesis , Protozoan Proteins/chemistry , Protozoan Proteins/genetics
14.
J Immunol ; 185(1): 551-9, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20525887

ABSTRACT

Dendritic cell (DC)-expressed CD40 is shown to play crucial roles in eliciting effector T cell responses, primarily the proinflammatory CD4(+) Th subsets and cytotoxic CD8(+) T cells that eliminate various infections and tumors, respectively. In contrast, DCs are also implied in the generation of regulatory T cells (Tregs) that counteract the functions of the proinflammatory Th subsets and exacerbate infections. However, the role of DC-expressed CD40 in the generation of Tregs is unknown. In this study, we generated bone marrow-derived DCs from mice (on a BALB/c background) expressing different levels of CD40 and tested their relative efficiency in generating Tregs. We observed that low levels of CD40 expression were required for efficient Treg generation. DCs expressing low levels of CD40 induced Tregs, whereas DCs expressing high levels of CD40 induced effector T cells, possibly CD8(+)CD40(+) T cells with a contraregulatory activity; the adoptive transfer of the former DC exacerbated whereas the latter significantly reduced Leishmania donovani infection in BALB/c mice. Similarly, priming of mice with leishmanial Ag-pulsed DCs expressing high levels of CD40 induced host protection against L. donovani challenge infection. In contrast, priming with the low CD40-expressing DC resulted in aggravated infection as compared with the control mice. The results establish that CD40 can play differential roles in Treg differentiation and determine the course of infection. We demonstrate that the knowledge can be efficiently used in adoptive cell transfer therapy against an infectious disease.


Subject(s)
Antigens, Protozoan/physiology , CD40 Antigens/physiology , Leishmania donovani/immunology , Leishmaniasis, Visceral/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/parasitology , Adoptive Transfer , Animals , Antigens, Protozoan/biosynthesis , Antigens, Protozoan/genetics , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Bone Marrow Cells/parasitology , Bone Marrow Transplantation/immunology , Bone Marrow Transplantation/pathology , CD40 Antigens/biosynthesis , CD40 Antigens/deficiency , Cells, Cultured , Coculture Techniques , Dendritic Cells/immunology , Dendritic Cells/parasitology , Dendritic Cells/transplantation , Genetic Predisposition to Disease , Immunophenotyping , Leishmaniasis, Visceral/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , T-Lymphocytes, Regulatory/metabolism
15.
J Biol Chem ; 285(32): 24855-62, 2010 Aug 06.
Article in English | MEDLINE | ID: mdl-20529864

ABSTRACT

Molecular interactions between the VAR2CSA protein, expressed on the surface of Plasmodium falciparum-infected erythrocytes, and placental chondroitin sulfate A (CSA) are primarily responsible for pregnancy-associated malaria (PAM). Interrupting these interactions may prevent or ameliorate the severity of PAM. Several of the Duffy binding-like (DBL) domains of VAR2CSA, including the DBL3x domain, have been shown to bind CSA in vitro, but a more detailed understanding of how DBL domains bind CSA is needed. In this study, we demonstrate that subdomain 3 (S3), one of the three subdomains of VAR2CSA DBL3x by itself, is the major contributor toward CSA binding. NMR spectroscopy and flow cytometry analyses show that S3 and the intact DBL3x domain bind CSA similarly. Mutations within the S3 portion of DBL3x markedly affect CSA binding. Both recombinant molecules, S3 and DBL3x, are recognized by antibodies in the plasma of previously pregnant women living in malaria-endemic regions of Mali, but much less so by plasma from men of the same regions. As the S3 sequence is highly conserved in all known VAR2CSA proteins expressed by different parasite isolates obtained from various malaria endemic areas of the world, the identification of S3 as an independent CSA-binding region provides a compelling molecular basis for designing interventions against PAM.


Subject(s)
Antigens, Protozoan/chemistry , Chondroitin Sulfates/chemistry , Plasmodium falciparum/metabolism , Animals , Antigens, Protozoan/physiology , CHO Cells , Cricetinae , Cricetulus , Erythrocytes/metabolism , Erythrocytes/parasitology , Female , Flow Cytometry/methods , Humans , Pregnancy , Pregnancy Complications, Parasitic , Protein Binding , Protein Folding
16.
Acta Trop ; 114(3): 131-7, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19913491

ABSTRACT

The intracellular trafficking of an Erythrocyte Binding Like (EBL) ligand has recently been shown to dramatically affect the multiplication rate and virulence of the rodent malaria parasite Plasmodium yoelii yoelii. In this review, we describe the current understanding of the role of EBL and other erythrocyte binding ligands in erythrocyte invasion, and discuss the mechanisms by which they may control multiplication rates and virulence in malaria parasites.


Subject(s)
Antigens, Protozoan/physiology , Erythrocytes/parasitology , Plasmodium/pathogenicity , Protozoan Proteins/physiology , Receptors, Cell Surface/physiology , Virulence Factors/physiology , Amino Acid Sequence , Animals , Humans , Models, Molecular , Molecular Sequence Data , Plasmodium/growth & development , Protein Structure, Tertiary , Protein Transport , Virulence
17.
Cell Microbiol ; 12(6): 725-39, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20039882

ABSTRACT

Parasite proteases play key roles in several fundamental steps of the Plasmodium life cycle, including haemoglobin degradation, host cell invasion and parasite egress. Plasmodium exit from infected host cells appears to be mediated by a class of papain-like cysteine proteases called 'serine repeat antigens' (SERAs). A SERA subfamily, represented by Plasmodium falciparum SERA5, contains an atypical active site serine residue instead of a catalytic cysteine. Members of this SERAser subfamily are abundantly expressed in asexual blood stages, rendering them attractive drug and vaccine targets. In this study, we show by antibody localization and in vivo fluorescent tagging with the red fluorescent protein mCherry that the two P. berghei serine-type family members, PbSERA1 and PbSERA2, display differential expression towards the final stages of merozoite formation. Via targeted gene replacement, we generated single and double gene knockouts of the P. berghei SERAser genes. These loss-of-function lines progressed normally through the parasite life cycle, suggesting a specialized, non-vital role for serine-type SERAs in vivo. Parasites lacking PbSERAser showed increased expression of the cysteine-type PbSERA3. Compensatory mechanisms between distinct SERA subfamilies may thus explain the absence of phenotypical defect in SERAser disruptants, and challenge the suitability to develop potent antimalarial drugs based on specific inhibitors of Plasmodium serine-type SERAs.


Subject(s)
Antigens, Protozoan/physiology , Plasmodium berghei/enzymology , Plasmodium berghei/growth & development , Protozoan Proteins/physiology , Serine Proteases/physiology , Amino Acid Sequence , Animals , Cell Line , Gene Expression Profiling , Gene Knockout Techniques , Humans , Life Cycle Stages , Malaria/parasitology , Mice , Microscopy, Fluorescence , Molecular Sequence Data , Parasitemia , Plasmodium berghei/pathogenicity , Rats , Rats, Sprague-Dawley
18.
Cell Microbiol ; 11(11): 1671-87, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19614665

ABSTRACT

The Apicomplexan parasite responsible for the most virulent form of malaria, Plasmodium falciparum, invades human erythrocytes through multiple ligand-receptor interactions. The P. falciparum reticulocyte-binding protein homologue (PfRh or PfRBL) family have been implicated in the invasion process but their exact role is unknown. PfRh1 and PfRh4, members of this protein family, bind to red blood cells and function in merozoite invasion during which they undergo a series of proteolytic cleavage events before and during entry into the host cell. The ectodomain of PfRh1 and PfRh4 are processed to produce fragments consistent with cleavage in the transmembrane domain and released into the supernatant, at about the time of invasion, in a manner consistent with rhomboid protease cleavage. Processing of both PfRh1 and PfRh4, and by extrapolation all membrane-bound members of this protein family, is important for function and release of these proteins on the merozoite surface and they along with EBA-175 are important components of the tight junction, the transient structure that links the erythrocyte via receptor-ligand interactions to the actin-myosin motor in the invading merozoite.


Subject(s)
Cell Adhesion Molecules/physiology , Cell Adhesion , Erythrocytes/parasitology , Membrane Proteins/physiology , Plasmodium falciparum/pathogenicity , Protozoan Proteins/physiology , Virulence Factors/physiology , Animals , Antigens, Protozoan/physiology , Humans , Models, Biological , Protein Processing, Post-Translational
19.
Infect Immun ; 77(9): 3857-63, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19546191

ABSTRACT

The Duffy binding-like (DBL) domains are common adhesion modules present in Plasmodium falciparum erythrocyte membrane protein 1 (PfEMP1) variants, which are responsible for immune evasion and cytoadherence. Knowledge about how immune responses are acquired against polymorphic DBL domains of PfEMP1 can aid in the development of vaccines for malaria. A recombinant DBLalpha domain, encoded by R29 var1, which binds complement receptor 1 to mediate rosetting by the P. falciparum laboratory strain R29, was expressed in Escherichia coli, renatured by oxidative refolding to its native form, and purified to homogeneity. Antibody levels in 704 plasmas obtained from residents of areas of different levels of malaria endemicity in Orissa (India) and Manhiça (Mozambique) were assessed by enzyme-linked immunosorbent assay. The refolded DBLalpha domain was pure, homogeneous, and functional in that it bound human erythrocytes with specificity and was capable of inhibiting rosetting. The proportion of individuals who had measurable anti-DBLalpha immunoglobulin G responses was low in areas of low malaria endemicity in Orissa (6.7%) but high in areas of high endemicity in Orissa (87.5%) and Manhiça (74.5%). Seroprevalence and antibody levels against the recombinant protein increased with the age of inhabitants from areas with high transmission rates (P < 0.001). Half of the children in these areas had seroconverted by the age of 5 years. These findings suggest that in spite of the extreme polymorphism of PfEMP1 DBLalpha domains, the acquisition of specific antibodies is rapid and age related and reflects the reduced risk of malaria in areas with high transmission rates. Further studies are required to elucidate the role of these antibodies in protection from malaria.


Subject(s)
Antigens, Protozoan/physiology , Protozoan Proteins/physiology , Receptors, Cell Surface/physiology , Rosette Formation , Animals , Antibodies, Protozoan/blood , Antigens, Protozoan/chemistry , Antigens, Protozoan/immunology , Enzyme-Linked Immunosorbent Assay , Humans , Immunoglobulin G/blood , Plasmodium falciparum , Protein Folding , Protein Structure, Tertiary , Protozoan Proteins/chemistry , Protozoan Proteins/immunology , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/immunology
20.
Proc Natl Acad Sci U S A ; 106(13): 5348-52, 2009 Mar 31.
Article in English | MEDLINE | ID: mdl-19279206

ABSTRACT

In the war against Plasmodium, humans have evolved to eliminate or modify proteins on the erythrocyte surface that serve as receptors for parasite invasion, such as the Duffy blood group, a receptor for Plasmodium vivax, and the Gerbich-negative modification of glycophorin C for Plasmodium falciparum. In turn, the parasite counters with expansion and diversification of ligand families. The high degree of polymorphism in glycophorin B found in malaria-endemic regions suggests that it also may be a receptor for Plasmodium, but, to date, none has been identified. We provide evidence from erythrocyte-binding that glycophorin B is a receptor for the P. falciparum protein EBL-1, a member of the Duffy-binding-like erythrocyte-binding protein (DBL-EBP) receptor family. The erythrocyte-binding domain, region 2 of EBL-1, expressed on CHO-K1 cells, bound glycophorin B(+) but not glycophorin B-null erythrocytes. In addition, glycophorin B(+) but not glycophorin B-null erythrocytes adsorbed native EBL-1 from the P. falciparum culture supernatants. Interestingly, the Efe pygmies of the Ituri forest in the Democratic Republic of the Congo have the highest gene frequency of glycophorin B-null in the world, raising the possibility that the DBL-EBP family may have expanded in response to the high frequency of glycophorin B-null in the population.


Subject(s)
Antigens, Protozoan/physiology , Erythrocytes/parasitology , Glycophorins/metabolism , Plasmodium falciparum/pathogenicity , Protozoan Proteins/physiology , Receptors, Cell Surface/physiology , Animals , Erythrocytes/metabolism , Glycophorins/genetics , Humans , Ligands , Molecular Sequence Data , Polymorphism, Genetic , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL
...