Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Molecules ; 19(9): 13990-4003, 2014 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-25197935

RESUMEN

A specific galactose-binding lectin was shown to inhibit the hemolytic effect of streptolysin O (SLO), an exotoxin produced by Streptococcus pyogenes. Commercially available lectins that recognize N-acetyllactosamine (ECA), T-antigen (PNA), and Tn-antigen (ABA) agglutinated rabbit erythrocytes, but had no effect on SLO-induced hemolysis. In contrast, SLO-induced hemolysis was inhibited by AKL, a lectin purified from sea hare (Aplysia kurodai) eggs that recognizes α-galactoside oligosaccharides. This inhibitory effect was blocked by the co-presence of d-galactose, which binds to AKL. A possible explanation for these findings is that cholesterol-enriched microdomains containing glycosphingolipids in the erythrocyte membrane become occupied by tightly stacked lectin molecules, blocking the interaction between cholesterol and SLO that would otherwise result in penetration of the membrane. Growth of S. pyogenes was inhibited by lectins from a marine invertebrate (AKL) and a mushroom (ABA), but was promoted by a plant lectin (ECA). Both these inhibitory and promoting effects were blocked by co-presence of galactose in the culture medium. Our findings demonstrate the importance of glycans and lectins in regulating mechanisms of toxicity, creation of pores in the target cell membrane, and bacterial growth.


Asunto(s)
Aplysia/química , Eritrocitos/fisiología , Galectinas/química , Hemólisis/efectos de los fármacos , Animales , Antibacterianos/química , Antibacterianos/aislamiento & purificación , Antibacterianos/farmacología , Proteínas Bacterianas/farmacología , Eritrocitos/efectos de los fármacos , Galectinas/aislamiento & purificación , Galectinas/farmacología , Hemolíticos/farmacología , Pruebas de Sensibilidad Microbiana , Óvulo/química , Conejos , Streptococcus pyogenes/efectos de los fármacos , Estreptolisinas/farmacología
2.
Parasitol Int ; 101: 102892, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38565335

RESUMEN

Reducing Plasmodium parasite transmission via the mosquito vector is a promising strategy for malaria control and elimination in endemic regions. In the mosquito midgut after the ingestion of an infected blood meal, malaria parasite gametes egress from erythrocytes and fertilize to develop into motile ookinetes that traverse midgut epithelial cells and transform into oocysts adjacent the basal lamina. Plasmodium ookinetes and young oocysts possess a unique organelle called the crystalloid; which has a honeycomb-like matrix structure and is indicated to be involved in sporozoite formation and maturation. In this study, we identified a novel crystalloid protein, PY17X_1113800, that is exclusively expressed in developing ookinetes. The protein possesses a signal peptide sequence, but lacks a transmembrane domain or GPI anchor signal sequence, as well as predicted adhesive domains which are characterisitic of many crystalloid proteins. The protein is highly conserved across the phylum Apicomplexa and within the greater clade Alveolata, such as Vitrella and the ciliates Paramecium and Tetrahymena, but is absent in cryptosporidia.


Asunto(s)
Proteínas Protozoarias , Proteínas Protozoarias/metabolismo , Proteínas Protozoarias/genética , Animales , Plasmodium , Oocistos , Orgánulos , Mosquitos Vectores/parasitología , Anopheles/parasitología
3.
Parasitol Int ; 100: 102864, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38331312

RESUMEN

Plasmodium falciparum accounts for the majority of malaria deaths, due to pathology provoked by the ability of infected erythrocytes to adhere to vascular endothelium within deep tissues. The parasite recognizes endothelium by trafficking and displaying protein ligands on the surface of asexual stage infected erythrocytes, such as members of the large family of pathogenic proteins, P. falciparum erythrocyte membrane protein 1 (PfEMP1). Parasite-encoded skeleton binding protein 1 (SBP1) plays an important role in the transport of these binding-related surface proteins, via cleft-like membranous structures termed Maurer's clefts, which are present within the cytoplasm of infected erythrocytes. Erythrocytes infected with gametocyte stages accumulate in the extravascular compartment of bone marrow; and it was suggested that their surface-expressed adhesion molecule profile and protein trafficking mechanisms might differ from those in asexual stage parasites. Protein trafficking mechanisms via Maurer's clefts have been well investigated in asexual stage parasite-infected erythrocytes; but little is known regarding the gametocyte stages. In this study, we characterized SBP1 during gametocyte maturation and demonstrated that SBP1 is expressed and localizes to dot-like Maurer's cleft structures in the cytoplasm of gametocyte-infected erythrocytes. Co-immunoprecipitation and mass spectrometry assays indicated that SBP1 interacts with the molecular chaperones PfHSP70-1 and PfHSP70-x. Localization analysis suggested that some PfHSP70-1 and/or PfHSP70-x localize in a dot-like pattern within the cytoplasm of immature gametocyte-infected erythrocytes. These findings suggest that SBP1 may interact with HSP70 chaperones in the infected erythrocyte cytoplasm during the immature gametocyte stages.


Asunto(s)
Proteínas Portadoras , Malaria Falciparum , Animales , Proteínas Portadoras/metabolismo , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/metabolismo , Eritrocitos/parasitología , Transporte de Proteínas , Malaria Falciparum/parasitología , Proteínas de la Membrana/metabolismo , Esqueleto/metabolismo
4.
Front Cell Infect Microbiol ; 13: 1197126, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37457963

RESUMEN

Plasmodium species cause malaria, and in the instance of Plasmodium falciparum is responsible for a societal burden of over 600,000 deaths annually. The symptoms and pathology of malaria are due to intraerythocytic parasites. Erythrocyte invasion is mediated by the parasite merozoite stage, and is accompanied by the formation of a parasitophorous vacuolar membrane (PVM), within which the parasite develops. The merozoite apical rhoptry organelle contains various proteins that contribute to erythrocyte attachment and invasion. RON3, a rhoptry bulb membrane protein, undergoes protein processing and is discharged into the PVM during invasion. RON3-deficient parasites fail to develop beyond the intraerythrocytic ring stage, and protein export into erythrocytes by the Plasmodium translocon of exported proteins (PTEX) apparatus is abrogated, as well as glucose uptake into parasites. It is known that truncated N- and C-terminal RON3 fragments are present in rhoptries, but it is unclear which RON3 fragments contribute to protein export by PTEX and glucose uptake through the PVM. To investigate and distinguish the roles of the RON3 C-terminal fragment at distinct developmental stages, we used a C-terminus tag for conditional and post-translational control. We demonstrated that RON3 is essential for blood-stage parasite survival, and knockdown of RON3 C-terminal fragment expression from the early schizont stage induces a defect in erythrocyte invasion and the subsequent development of ring stage parasites. Protein processing of full-length RON3 was partially inhibited in the schizont stage, and the RON3 C-terminal fragment was abolished in subsequent ring-stage parasites compared to the RON3 N-terminal fragment. Protein export and glucose uptake were abrogated specifically in the late ring stage. Plasmodial surface anion channel (PSAC) activity was partially retained, facilitating small molecule traffic across the erythrocyte membrane. The knockdown of the RON3 C-terminal fragment after erythrocyte invasion did not alter parasite growth. These data suggest that the RON3 C-terminal fragment participates in erythrocyte invasion and serves an essential role in the progression of ring-stage parasite growth by the establishment of the nutrient-permeable channel in the PVM, accompanying the transport of ring-stage parasite protein from the plasma membrane to the PVM.


Asunto(s)
Malaria , Parásitos , Plasmodium , Animales , Plasmodium falciparum/genética , Parásitos/metabolismo , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Transporte de Proteínas , Eritrocitos/parasitología , Plasmodium/metabolismo , Glucosa/metabolismo , Proliferación Celular
5.
Biomolecules ; 13(3)2023 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-36979393

RESUMEN

Plasmodium malaria parasites use erythrocyte-binding-like (EBL) ligands to invade erythrocytes in their vertebrate host. EBLs are released from micronemes, which are secretory organelles located at the merozoite apical end and bind to erythrocyte surface receptors. Because of their essential nature, EBLs have been studied as vaccine candidates, such as the Plasmodium vivax Duffy binding protein. Previously, we showed through using the rodent malaria parasite Plasmodium yoelii that a single amino acid substitution within the EBL C-terminal Cys-rich domain (region 6) caused mislocalization of this molecule and resulted in alteration of the infection course and virulence between the non-lethal 17X and lethal 17XL strains. In the present study, we generated a panel of transgenic P. yoelii lines in which seven of the eight conserved Cys residues in EBL region 6 were independently substituted to Ala residues to observe the consequence of these substitutions with respect to EBL localization, the infection course, and virulence. Five out of seven transgenic lines showed EBL mislocalizations and higher parasitemias. Among them, three showed increased virulence, whereas the other two did not kill the infected mice. The remaining two transgenic lines showed low parasitemias similar to their parental 17X strain, and their EBL localizations did not change. The results indicate the importance of Cys residues in EBL region 6 for EBL localization, parasite infection course, and virulence and suggest an association between EBL localization and the parasite infection course.


Asunto(s)
Malaria , Plasmodium yoelii , Animales , Ratones , Ligandos , Cisteína/metabolismo , Plasmodium yoelii/genética , Plasmodium yoelii/metabolismo , Parasitemia , Secuencia de Aminoácidos , Proteínas Protozoarias/metabolismo , Moléculas de Adhesión Celular/metabolismo , Malaria/metabolismo , Eritrocitos/metabolismo
6.
Proc Natl Acad Sci U S A ; 106(17): 7167-72, 2009 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-19346470

RESUMEN

The major virulence determinant of the rodent malaria parasite, Plasmodium yoelii, has remained unresolved since the discovery of the lethal line in the 1970s. Because virulence in this parasite correlates with the ability to invade different types of erythrocytes, we evaluated the potential role of the parasite erythrocyte binding ligand, PyEBL. We found 1 amino acid substitution in a domain responsible for intracellular trafficking between the lethal and nonlethal parasite lines and, furthermore, that the intracellular localization of PyEBL was distinct between these lines. Genetic modification showed that this substitution was responsible not only for PyEBL localization but also the erythrocyte-type invasion preference of the parasite and subsequently its virulence in mice. This previously unrecognized mechanism for altering an invasion phenotype indicates that subtle alterations of a malaria parasite ligand can dramatically affect host-pathogen interactions and malaria virulence.


Asunto(s)
Aminoácidos/metabolismo , Eritrocitos/metabolismo , Plasmodium yoelii/metabolismo , Plasmodium yoelii/patogenicidad , Proteínas Protozoarias/metabolismo , Secuencia de Aminoácidos , Aminoácidos/genética , Animales , Ligandos , Ratones , Microscopía Inmunoelectrónica , Datos de Secuencia Molecular , Plasmodium yoelii/genética , Proteínas Protozoarias/química , Proteínas Protozoarias/genética , Virulencia
8.
Parasitol Int ; 84: 102407, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34147682

RESUMEN

Plasmodium parasites cause malaria in mammalian hosts and are transmitted by Anopheles mosquitoes. Activated gametocytes in the mosquito midgut egress from erythrocytes followed by fertilization and zygote formation. Zygotes differentiate into motile invasive ookinetes, which penetrate the midgut epithelium before forming oocysts beneath the basal lamina. Ookinete development and traversal across the mosquito midgut wall are major bottlenecks in the parasite life cycle. In ookinetes, surface proteins and proteins stored in apical organelles have been shown to be involved in parasite-host interactions. A group of ookinete proteins that are predicted to have such functions are named PSOPs (putative secreted ookinete protein). PSOP1 is possibly involved in migration through the midgut wall, and here its subcellular localization was examined in ookinetes by immunoelectron microscopy. PSOP1 localizes to the micronemes of Plasmodium yoelii and Plasmodium berghei ookinetes, indicating that it is stored and possibly apically secreted during ookinete penetration through the mosquito midgut wall.


Asunto(s)
Malaria/parasitología , Plasmodium berghei/genética , Plasmodium yoelii/genética , Proteínas Protozoarias/genética , Animales , Interacciones Huésped-Parásitos , Ratones , Oocistos/fisiología , Orgánulos/parasitología , Proteínas Protozoarias/metabolismo
9.
Mol Biochem Parasitol ; 236: 111261, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31981605

RESUMEN

Plasmodium parasites cause malaria in mammalian hosts and are transmitted by Anopheles mosquitoes. Gametocytes, which differentiate from asexual-stage parasites, are activated by environmental changes when ingested into the mosquito midgut, and are rapidly released from erythrocytes prior to fertilization. Secretory proteins localized to osmiophilic bodies (OBs), organelles unique to gametocytes, have been reported to be involved in female gametocyte egress. In this study, we investigate the dynamics of OBs in activated gametocytes of Plasmodium falciparum and Plasmodium yoelii using the female OB-specific marker protein, G377. After activation, female gametocyte OBs migrate to the parasite surface and fuse to form large vesicles beneath the parasite plasma membrane. At the marginal region of female gametocytes, fused vesicles secrete contents by exocytosis into the parasitophorous vacuole space, prior to parasite egress via the break-down of the erythrocyte membrane. This is the first detailed description of how proteins are transported through osmiophilic bodies.


Asunto(s)
Plasmodium falciparum , Plasmodium yoelii , Proteínas Protozoarias/ultraestructura , Eritrocitos/parasitología , Malaria/parasitología , Microscopía Inmunoelectrónica/métodos , Orgánulos/metabolismo , Orgánulos/ultraestructura , Plasmodium falciparum/metabolismo , Plasmodium falciparum/ultraestructura , Plasmodium yoelii/metabolismo , Plasmodium yoelii/ultraestructura , Proteínas Protozoarias/metabolismo
10.
Parasitol Int ; 75: 102003, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31669509

RESUMEN

Plasmodium falciparum proteins involved in vascular endothelial cell adherence are transported to the surface of infected erythrocytes. These proteins are exported through parasite-derived membrane structures within the erythrocyte cytoplasm called Maurer's clefts. Skeleton binding protein 1 (SBP1) is localized in the Maurer's clefts and plays an important role in transporting molecules to the surface of infected erythrocytes. Details of the translocation pathway are unclear and in this study we focused on the subcellular localization of SBP1 at an early intraerythrocytic stage. We performed immunoelectron microscopy using specific anti-SBP1 antibodies generated by immunization with recombinant SBP1 of P. falciparum. At the early trophozoite (ring form) stage, SBP1 was detected within an electron dense material (EDM) found in the parasite cytoplasm and in the parasitophorous vacuolar (PV) space. These findings demonstrate that SBP1 accumulates in EDM in the early trophozoite cytoplasm and is transported to the PV space before translocation to the Maurer's clefts formed in the erythrocyte cytoplasm.


Asunto(s)
Eritrocitos/parasitología , Proteínas de la Membrana/genética , Plasmodium falciparum/genética , Proteínas Protozoarias/genética , Vacuolas/parasitología , Animales , Membrana Celular/parasitología , Citoplasma/parasitología , Proteínas de la Membrana/metabolismo , Microscopía Inmunoelectrónica , Plasmodium falciparum/metabolismo , Transporte de Proteínas , Proteínas Protozoarias/metabolismo , Conejos
11.
Parasitol Int ; 68(1): 17-23, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30290224

RESUMEN

Invasion of host cells by apicomplexan parasites is mediated by proteins released from microneme, rhoptry, and dense granule secretory organelles located at the apical end of parasite invasive forms. Microneme secreted proteins establish interactions with host cell receptors and induce exocytosis of the rhoptry organelle. Rhoptry proteins are involved in target cell invasion as well as the formation of the parasitophorous vacuole in which parasites reside during development within the host cell. In Plasmodium merozoites, the rhoptry neck protein (RON) complex consists of RON2, RON4, and RON5, and interacts with apical membrane antigen 1 (AMA1) as a critical structure of the invasion moving junction. PfRON12 is known to localize to the rhoptry neck of merozoites, but its function remains obscure. The roles of RON proteins are largely unknown in sporozoites, the second invasive form of Plasmodium which possesses a conserved apical end secretory structure. Here, we confirm that RON12 is expressed in the rhoptry neck of merozoites in rodent malaria parasites, whereas in contrast we show that RON12 is localized to the rhoptry body in sporozoites. Phenotypic analysis of Plasmodium berghei ron12-disrupted mutants revealed that RON12 is dispensable for sporogony, invasion of mosquito salivary glands and mouse hepatocytes, and development in hepatocytes.


Asunto(s)
Interacciones Huésped-Parásitos/genética , Plasmodium/genética , Proteínas Protozoarias/genética , Esporozoítos/genética , Animales , Proteínas Portadoras , Eritrocitos/parasitología , Femenino , Eliminación de Gen , Células Hep G2 , Hepatocitos/parasitología , Humanos , Merozoítos/genética , Merozoítos/metabolismo , Ratones , Orgánulos/metabolismo , Plasmodium/crecimiento & desarrollo , Plasmodium/inmunología , Plasmodium/fisiología , Plasmodium berghei/genética , Plasmodium berghei/crecimiento & desarrollo , Plasmodium berghei/fisiología , Proteínas Protozoarias/inmunología , Esporozoítos/crecimiento & desarrollo , Esporozoítos/fisiología
12.
Parasitol Int ; 68(1): 87-91, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30342119

RESUMEN

Proteins coating Plasmodium merozoite surface and secreted from its apical organelles are considered as promising vaccine candidates for blood-stage malaria. The rhoptry neck protein 12 of Plasmodium falciparum (PfRON12) was recently reported as a protein specifically expressed in schizonts and localized to the rhoptry neck of merozoites. Here, we assessed its potential as a vaccine candidate. We expressed a recombinant PfRON12 protein by a wheat germ cell-free system to obtain anti-PfRON12 antibody. Immunoblot analysis of schizont lysates detected a single band at approximately 40 kDa under reducing conditions, consistent with the predicted molecular weight. Additionally, anti-PfRON12 antibody recognized a single band around 80 kDa under non-reducing conditions, suggesting native PfRON12 forms a disulfide-bond-mediated multimer. Immunofluorescence assay and immunoelectron microscopy revealed that PfRON12 localized to the rhoptry neck of merozoites in schizonts and to the surface of free merozoites. The biological activity of anti-PfRON12 antibody was tested by in vitro growth inhibition assay (GIA), and the rabbit antibodies significantly inhibited merozoite invasion of erythrocytes. We then investigated whether PfRON12 is immunogenic in P. falciparum-infected individuals. The sera from P. falciparum infected individuals in Thailand and Mali reacted with the recombinant PfRON12. Furthermore, human anti-PfRON12 antibodies affinity-purified from Malian serum samples inhibited merozoite invasion of erythrocytes in vitro. Moreover, pfron12 is highly conserved with only 4 non-synonymous mutations in the coding sequence from approximately 200 isolates deposited in PlasmoDB. These results suggest that PfRON12 might be a potential blood-stage vaccine candidate antigen against P. falciparum.


Asunto(s)
Antígenos de Protozoos/inmunología , Eritrocitos/parasitología , Merozoítos/fisiología , Plasmodium falciparum/inmunología , Proteínas Protozoarias/inmunología , Proteínas Tirosina Quinasas Receptoras/inmunología , Animales , Anticuerpos Antiprotozoarios/sangre , Antígenos de Protozoos/genética , Infecciones Asintomáticas/epidemiología , Ensayo de Inmunoadsorción Enzimática , Eritrocitos/inmunología , Técnica del Anticuerpo Fluorescente , Humanos , Vacunas contra la Malaria/genética , Vacunas contra la Malaria/inmunología , Malaria Falciparum/epidemiología , Malaria Falciparum/inmunología , Malaria Falciparum/parasitología , Malaria Falciparum/prevención & control , Malí/epidemiología , Merozoítos/inmunología , Conejos , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Esquizontes/química , Tailandia/epidemiología
13.
Infect Immun ; 76(4): 1702-8, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18268027

RESUMEN

One of the major bottlenecks in malaria research has been the difficulty in recombinant protein expression. Here, we report the application of the wheat germ cell-free system for the successful production of malaria proteins. For proof of principle, the Pfs25, PfCSP, and PfAMA1 proteins were chosen. These genes contain very high A/T sequences and are also difficult to express as recombinant proteins. In our wheat germ cell-free system, native and codon-optimized versions of the Pfs25 genes produced equal amounts of proteins. PfCSP and PfAMA1 genes without any codon optimization were also expressed. The products were soluble, with yields between 50 and 200 mug/ml of the translation mixture, indicating that the cell-free system can be used to produce malaria proteins without any prior optimization of their biased codon usage. Biochemical and immunocytochemical analyses of antibodies raised in mice against each protein revealed that every antibody retained its high specificity to the parasite protein in question. The development of parasites in mosquitoes fed patient blood carrying Plasmodium falciparum gametocytes and supplemented with our mouse anti-Pfs25 sera was strongly inhibited, indicating that both Pfs25-3D7/WG and Pfs25-TBV/WG retained their immunogenicity. Lastly, we carried out a parallel expression assay of proteins of blood-stage P. falciparum. The PCR products of 124 P. falciparum genes chosen from the available database were used directly in a small-scale format of transcription and translation reactions. Autoradiogram testing revealed the production of 93 proteins. The application of this new cell-free system-based protocol for the discovery of malaria vaccine candidates will be discussed.


Asunto(s)
Sistema Libre de Células/química , Vacunas contra la Malaria , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/química , Proteínas Recombinantes , Triticum , Animales , Antígenos de Protozoos , Culicidae , Humanos , Ratones , Biosíntesis de Proteínas , Vacunas Sintéticas
14.
Mol Biochem Parasitol ; 158(1): 11-21, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18155305

RESUMEN

A complex of high-molecular-mass proteins (PfRhopH) of the human malaria parasite Plasmodium falciparum induces host protective immunity and therefore is a candidate for vaccine development. Understanding the level of polymorphism and the evolutionary processes is important for advancements in both vaccine design and knowledge of the evolution of cell invasion in this parasite. In the present study, we sequenced the entire open reading frames of seven genes encoding the proteins of the PfRhopH complex (rhoph2, rhoph3, and five rhoph1/clag gene paralogs). We found that four rhoph1/clag genes (clag2, 3.1, 3.2, and 8) were highly polymorphic. Amino acid substitutions and indels are predominantly clustered around amino acid positions 1000-1200 of these four rhoph1/clag genes. An excess of nonsynonymous substitutions over synonymous substitutions was detected for clag8 and 9, indicating positive selection. The McDonald-Kreitman test with a Plasmodium reichenowi orthologous sequence also supports positive selection on clag8. Based on the ratio of interspecific genetic distance to intraspecific distance, the time to the most recent common ancestor of the clag2 and 8 polymorphisms was estimated to be 1.89 and 0.87 million years ago, respectively, assuming divergence of P. falciparum and P. reichenowi 6 million years ago. In addition to a copy number polymorphism, gene conversion events were detected for the rhoph1/clag genes on chromosome 3, which likely play a role in increasing the diversity of each locus. Our results indicate that a high diversity of the PfRhopH1/Clag multigene family is maintained by diversifying selection forces over a considerably long period.


Asunto(s)
Evolución Molecular , Plasmodium falciparum/genética , Polimorfismo Genético , Proteínas Protozoarias/genética , Secuencia de Aminoácidos , Sustitución de Aminoácidos/genética , Animales , Secuencia de Bases , ADN Protozoario/química , ADN Protozoario/genética , Conversión Génica , Dosificación de Gen , Mutación INDEL , Datos de Secuencia Molecular , Filogenia , Selección Genética , Alineación de Secuencia , Análisis de Secuencia de ADN , Homología de Secuencia de Aminoácido
15.
Parasitol Int ; 67(5): 637-639, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29886341

RESUMEN

Apical organellar proteins in Plasmodium falciparum merozoites play important roles upon invasion. To date, dense granule, the least studied apical organelle, secretes parasite proteins across the parasitophorous vacuole membrane (PVM) to remodel the infected erythrocyte. Although this phenomenon is key to parasite growth and virulence, only five proteins so far have been identified as dense granule proteins. Further elucidation of dense granule molecule(s) is therefore required. P. falciparum Exported Protein (EXP) 1, previously reported as a parasitophorous vacuole membrane (PVM) protein, is considered essential for parasite growth. In this study, we characterized EXP1 using specific anti-EXP1 antibodies generated by immunization of wheat germ cell-free produced recombinant EXP1. Immunofluorescence microscopy (IFA) demonstrated that EXP1 co-localized with RESA, indicating that the protein is initially localized to dense granules in merozoites, followed by translocation to the PVM. The EXP1 localization in dense granule of merozoites and its translocation to the PVM after invasion of erythrocytes were further confirmed by immunoelectron microscopy. Here, we demonstrate that EXP1 is one of the dense granule proteins in merozoites, which is then transported to the PVM after invasion.


Asunto(s)
Antígenos de Protozoos/metabolismo , Merozoítos/ultraestructura , Plasmodium falciparum/metabolismo , Antígenos de Protozoos/genética , Transporte Biológico , Eritrocitos/parasitología , Malaria Falciparum/parasitología , Microscopía Electrónica , Microscopía Fluorescente , Plasmodium falciparum/genética , Vacuolas/metabolismo
16.
Parasitol Int ; 55(3): 227-31, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16822707

RESUMEN

The Plasmodium circumsporozoite protein/thrombospondin-related anonymous protein-related protein (CTRP) is expressed at the mosquito midgut ookinete stage and is considered to be a transmission-blocking vaccine candidate. CTRP is composed of multiple von Willebrand factor A (vWA) and thrombospondin type 1 domains in the extracellular portion of the molecule, and a short acidic cytoplasmic domain that interacts with the actomyosin machinery. As a means to predict functionally relevant domains within CTRP we determined the nucleotide sequences of CTRP from the Plasmodium vivax Sall and the Plasmodium yoelii 17XL strains and characterized the conservation of domain architectures and motifs across Plasmodium genera. Sequence alignments indicate that the CTRP 1st to 4th vWA domains exhibit greater conservation, and thereby are perhaps functionally more important than the 5th and 6th domains. This point should be considered for the development of a transmission-blocking vaccine that includes CTRP recombinant subunit. To complement previous cellular studies on CTRP, we further determined the expression and cellular localization of CTRP protein in P. vivax and P. yoelii.


Asunto(s)
Plasmodium vivax/química , Proteínas Protozoarias/química , Receptores de Superficie Celular/química , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Estadios del Ciclo de Vida , Datos de Secuencia Molecular , Plasmodium gallinaceum/ultraestructura , Plasmodium vivax/ultraestructura , Plasmodium yoelii/química , Estructura Terciaria de Proteína , Proteínas Protozoarias/análisis , Proteínas Protozoarias/genética , Receptores de Superficie Celular/análisis , Receptores de Superficie Celular/genética , Alineación de Secuencia
17.
Mol Biochem Parasitol ; 143(1): 20-8, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15953647

RESUMEN

The Plasmodium falciparum high molecular mass rhoptry protein ('PfRhopH') complex is important for parasite growth and comprises three distinct gene products: RhopH1, RhopH2 and RhopH3. We have previously shown that P. falciparum RhopH1 is encoded by either PFC0110w (clag3.2) or PFC0120w (clag3.1), members of the previously-named clag (cytoadherence-linked asexual gene) multigene family. In this report, we have further characterized rhoph1/clag members in terms of gene structure, transcription and protein expression. The cDNA sequences for all five rhoph1/clag members were determined, confirming previous in silico predictions of intron-exon boundaries. All member genes were transcribed in HB3 and 3D7 parasite lines, but clag3.2 was not transcribed in Dd2 parasites. The peak abundance of transcripts for all genes was observed during the late schizont stage. Antisera specific to Clag2 and Clag3.1 localized these proteins to the apical end of merozoites in segmented schizonts, and both proteins are found to be components of the PfRhopH complex. PfRhopH complex that was immunoprecipitated with anti-Clag9 antibody contained neither Clag2 nor Clag3.1, thereby suggesting that PfRhopH complexes contain only individual rhoph1/clag gene products. Since the PfRhopH complex binds the erythrocyte surface, and RhopH2 and RhopH3 are encoded by single copy genes, the RhopH1/Clag proteins may serve to confer some degree of specificity to the roles of the individual complexes.


Asunto(s)
Plasmodium falciparum/genética , Proteínas Protozoarias/genética , Animales , Secuencia de Bases , Mapeo Cromosómico , Clonación Molecular , Cruzamientos Genéticos , ADN Protozoario/genética , Sustancias Macromoleculares
18.
Vaccine ; 33(16): 1901-8, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25765968

RESUMEN

Malaria transmission-blocking vaccines (TBV) aim to interfere with the development of the malaria parasite in the mosquito vector, and thus prevent spread of transmission in a community. To date three TBV candidates have been identified in Plasmodium vivax; namely, the gametocyte/gamete protein Pvs230, and the ookinete surface proteins Pvs25 and Pvs28. The Plasmodium falciparum gametocyte/gamete stage proteins Pfs48/45 and Pfs47 have been studied as TBV candidates, and Pfs48/45 shown to induce transmission-blocking antibodies, but the candidacy of their orthologs in P. vivax, Pvs48/45 (PVX_083235) and Pvs47 (PVX_083240), for vivax TBV have not been tested. Herein we investigated whether targeting Pvs48/45 and Pvs47 can inhibit parasite transmission to mosquitoes, using P. vivax isolates obtained in Thailand. Mouse antisera directed against the products from plasmids expressing Pvs48/45 and Pvs47 detected proteins of approximately 45- and 40-kDa, respectively, in the P. vivax gametocyte lysate, by Western blot analysis under non-reducing conditions. In immunofluorescence assays Pvs48/45 was detected predominantly on the surface and Pvs47 was detected in the cytoplasm of gametocytes. Membrane feeding transmission assays demonstrated that anti-Pvs48/45 and -Pvs47 mouse sera significantly reduced the number of P. vivax oocysts developing in the mosquito midgut. Limited amino acid polymorphism of these proteins was observed among 27 P. vivax isolates obtained from Thailand, Vanuatu, and Colombia; suggesting that polymorphism may not be an impediment for the utilization of Pvs48/45 and Pvs47 as TBV antigens. In one Thai isolate we found that the fourth cysteine residue in the Pvs47 cysteine-rich domain (CRD) III (amino acid position 337) is substituted to phenylalanine. However, antibodies targeting Pvs47 CRDI-III showed a significant transmission-reducing activity against this isolate, suggesting that this substitution in Pvs47 was not critical for recognition by the generated antibodies. In conclusion, our results indicate that Pvs48/45 and Pvs47 are potential transmission-blocking vaccine candidates of P. vivax.


Asunto(s)
Anticuerpos Antiprotozoarios/inmunología , Antígenos de Protozoos/inmunología , Vacunas contra la Malaria/inmunología , Malaria Vivax/prevención & control , Plasmodium vivax/inmunología , Vacunas de ADN/inmunología , Sustitución de Aminoácidos , Animales , Antígenos de Protozoos/química , Antígenos de Protozoos/genética , Femenino , Humanos , Vacunas contra la Malaria/administración & dosificación , Vacunas contra la Malaria/genética , Malaria Vivax/transmisión , Ratones , Péptidos/genética , Péptidos/inmunología , Plasmodium vivax/genética , Polimorfismo Genético , Ratas , Vacunación , Vacunas de ADN/administración & dosificación
19.
PLoS One ; 10(8): e0136109, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26292257

RESUMEN

BACKGROUND: Nearly 100% protection against malaria infection can be achieved in humans by immunization with P. falciparum radiation-attenuated sporozoites (RAS). Although it is thought that protection is mediated by T cell and antibody responses, only a few of the many pre-erythrocytic (sporozoite and liver stage) antigens that are targeted by these responses have been identified. METHODOLOGY: Twenty seven P. falciparum pre-erythrocytic antigens were selected using bioinformatics analysis and expression databases and were expressed in a wheat germ cell-free protein expression system. Recombinant proteins were recognized by plasma from RAS-immunized subjects, and 21 induced detectable antibody responses in mice and rabbit and sera from these immunized animals were used to characterize these antigens. All 21 proteins localized to the sporozoite: five localized to the surface, seven localized to the micronemes, cytoplasm, endoplasmic reticulum or nucleus, two localized to the surface and cytoplasm, and seven remain undetermined. PBMC from RAS-immunized volunteers elicited positive ex vivo or cultured ELISpot responses against peptides from 20 of the 21 antigens. CONCLUSIONS: These T cell and antibody responses support our approach of using reagents from RAS-immunized subjects to screen potential vaccine antigens, and have led to the identification of a panel of novel P. falciparum antigens. These results provide evidence to further evaluate these antigens as vaccine candidates. TRIAL REGISTRATION: ClinicalTrials.gov NCT00870987 ClinicalTrials.gov NCT00392015.


Asunto(s)
Antígenos de Protozoos/inmunología , Eritrocitos/inmunología , Vacunas contra la Malaria/inmunología , Malaria Falciparum/inmunología , Malaria Falciparum/prevención & control , Plasmodium falciparum/inmunología , Animales , Eritrocitos/parasitología , Humanos , Inmunización , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/parasitología , Vacunas contra la Malaria/farmacología , Malaria Falciparum/sangre , Ratones , Ratones Endogámicos BALB C , Proteínas Protozoarias/inmunología , Conejos , Esporozoítos/inmunología , Linfocitos T/inmunología , Linfocitos T/parasitología
20.
Jpn J Infect Dis ; 65(2): 152-6, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22446123

RESUMEN

We identified naturally induced antibodies from malaria patients in Thailand and clarified the effect of the antibodies on gametocyte development. Fifty-nine percent of the Plasmodium falciparum-infected blood samples (17 of 29) fed to female Anopheles mosquitoes showed no oocyst infection. Seventeen percent of the samples (5 of 29) distorted the morphology and hampered the maturity of the gametocytes. A possible mechanism for the gametocyte inhibitory activity was shown by the binding of the plasma antibodies to live, immature, intraerythrocytic gametocytes during the incubation period. One hundred fifty-seven proteins specific to different gametocyte stages were explored to find the targets of the antisera that bound to the live gametocytes. However, no additional gametocyte transmission-blocking vaccine candidate was detected. Therefore, the development of alternative transmission-blocking vaccines in high-transmission areas should focus on the identification of more gametocyte antigens-inducing inhibitory antibodies that reduce gametocytemia.


Asunto(s)
Anticuerpos Antiprotozoarios/sangre , Malaria Falciparum/inmunología , Plasmodium falciparum/inmunología , Animales , Anopheles/parasitología , Sangre/inmunología , Femenino , Humanos , Plasmodium falciparum/citología , Tailandia
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda