Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Parasitol Int ; 101: 102892, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38565335

RESUMO

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.


Assuntos
Proteínas de Protozoários , Proteínas de Protozoários/metabolismo , Proteínas de Protozoários/genética , Animais , Plasmodium , Oocistos , Organelas , Mosquitos Vetores/parasitologia , Anopheles/parasitologia
2.
Parasitol Int ; 100: 102864, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38331312

RESUMO

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.


Assuntos
Proteínas de Transporte , Malária Falciparum , Animais , Proteínas de Transporte/metabolismo , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/metabolismo , Eritrócitos/parasitologia , Transporte Proteico , Malária Falciparum/parasitologia , Proteínas de Membrana/metabolismo , Esqueleto/metabolismo
3.
Front Cell Infect Microbiol ; 13: 1197126, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37457963

RESUMO

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.


Assuntos
Malária , Parasitos , Plasmodium , Animais , Plasmodium falciparum/genética , Parasitos/metabolismo , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Transporte Proteico , Eritrócitos/parasitologia , Plasmodium/metabolismo , Glucose/metabolismo , Proliferação de Células
4.
mSphere ; 8(4): e0058722, 2023 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-37272704

RESUMO

During invasion, Plasmodium parasites secrete proteins from rhoptry and microneme apical end organelles, which have crucial roles in attaching to and invading target cells. A sporozoite stage-specific gene silencing system revealed that rhoptry neck protein 2 (RON2), RON4, and RON5 are important for sporozoite invasion of mosquito salivary glands. Here, we further investigated the roles of RON4 during sporozoite infection of the liver in vivo. Following intravenous inoculation of RON4-knockdown sporozoites into mice, we demonstrated that sporozoite RON4 has multiple functions during sporozoite traversal of sinusoidal cells and infection of hepatocytes. In vitro infection experiments using a hepatoma cell line revealed that secreted RON4 is involved in sporozoite adhesion to hepatocytes and has an important role in the early steps of hepatocyte infection. In addition, in vitro motility assays indicated that RON4 is required for sporozoite attachment to the substrate and the onset of migration. These findings indicate that RON4 is crucial for sporozoite migration toward and invasion of hepatocytes via attachment ability and motility.IMPORTANCEMalarial parasite transmission to mammals is established when sporozoites are inoculated by mosquitoes and migrate through the bloodstream to infect hepatocytes. Many aspects of the molecular mechanisms underpinning migration and cellular invasion remain largely unelucidated. By applying a sporozoite stage-specific gene silencing system in the rodent malarial parasite, Plasmodium berghei, we demonstrated that rhoptry neck protein 4 (RON4) is crucial for sporozoite infection of the liver in vivo. Combined with in vitro investigations, it was revealed that RON4 functions during a crossing of the sinusoidal cell layer and invading hepatocytes, at an early stage of liver infection, by mediating the sporozoite capacity for adhesion and the onset of motility. Since RON4 is also expressed in Plasmodium merozoites and Toxoplasma tachyzoites, our findings contribute to understanding the conserved invasion mechanisms of Apicomplexa parasites.


Assuntos
Malária , Plasmodium berghei , Esporozoítos , Animais , Camundongos , Plasmodium berghei/crescimento & desenvolvimento , Plasmodium berghei/fisiologia , Fígado/metabolismo , Fígado/parasitologia , Fígado/patologia , Malária/metabolismo , Malária/parasitologia , Malária/patologia , Esporozoítos/fisiologia , Proteínas de Protozoários/metabolismo , Hepatócitos/metabolismo , Hepatócitos/parasitologia , Hepatócitos/patologia
5.
Biomolecules ; 13(3)2023 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-36979393

RESUMO

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.


Assuntos
Malária , Plasmodium yoelii , Animais , Camundongos , Ligantes , Cisteína/metabolismo , Plasmodium yoelii/genética , Plasmodium yoelii/metabolismo , Parasitemia , Sequência de Aminoácidos , Proteínas de Protozoários/metabolismo , Moléculas de Adesão Celular/metabolismo , Malária/metabolismo , Eritrócitos/metabolismo
6.
Commun Biol ; 5(1): 773, 2022 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-35915227

RESUMO

A vaccine targeting multiple stages of the Plasmodium falciparum parasite life cycle is desirable. The sporozoite surface Circumsporozoite Protein (CSP) is the target of leading anti-infective P. falciparum pre-erythrocytic vaccines. Pfs230, a sexual-stage P. falciparum surface protein, is currently in trials as the basis for a transmission-blocking vaccine, which inhibits parasite development in the mosquito vector. Here, recombinant full-length CSP and a Pfs230 fragment (Pfs230D1+) are co-displayed on immunogenic liposomes to induce immunity against both infection and transmission. Liposomes contain cobalt-porphyrin phospholipid (CoPoP), monophosphoryl lipid A and QS-21, and rapidly bind His-tagged CSP and Pfs230D1+ upon admixture to form bivalent particles that maintain reactivity with conformational monoclonal antibodies. Use of multicolor fluorophore-labeled antigens reveals liposome binding upon admixture, stability in serum and enhanced uptake in murine macrophages in vitro. Bivalent liposomes induce humoral and cellular responses against both CSP and Pfs230D1+. Vaccine-induced antibodies reduce parasite numbers in mosquito midguts in a standard membrane feeding assay. Mice immunized with liposome-displayed antigens or that passively receive antibodies from immunized rabbits have reduced parasite liver burden following challenge with transgenic sporozoites expressing P. falciparum CSP.


Assuntos
Vacinas Antimaláricas , Plasmodium falciparum , Animais , Anticorpos Antiprotozoários , Antígenos , Lipossomos , Camundongos , Proteínas de Protozoários/genética , Coelhos , Esporozoítos
7.
Parasitol Int ; 87: 102525, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-34896614

RESUMO

Existing control measures have significantly reduced malaria morbidity and mortality in the last two decades, although these reductions are now stalling. Significant efforts have been undertaken to develop malaria vaccines. Recently, extensive progress in malaria vaccine development has been made for Plasmodium falciparum. To date, only the RTS,S/AS01 vaccine has been tested in Phase 3 clinical trials and is now under implementation, despite modest efficacy. Therefore, the development of a malaria transmission-blocking vaccine (TBV) will be essential for malaria elimination. Only a limited number of TBVs have reached pre-clinical or clinical development with several major challenges impeding their development, including low immunogenicity in humans. TBV development efforts against P. vivax, the second major cause of malaria morbidity, lag far behind those for P. falciparum. In this review we summarize the latest progress, challenges and innovations in P. vivax TBV research and discuss how to accelerate its development.


Assuntos
Vacinas Antimaláricas , Malária Vivax/prevenção & controle , Plasmodium vivax/imunologia , Humanos , Malária Falciparum/epidemiologia , Malária Falciparum/prevenção & controle , Malária Vivax/epidemiologia , Malária Vivax/transmissão , Plasmodium falciparum/imunologia , Desenvolvimento de Vacinas
8.
Parasitol Int ; 84: 102407, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34147682

RESUMO

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.


Assuntos
Malária/parasitologia , Plasmodium berghei/genética , Plasmodium yoelii/genética , Proteínas de Protozoários/genética , Animais , Interações Hospedeiro-Parasita , Camundongos , Oocistos/fisiologia , Organelas/parasitologia , Proteínas de Protozoários/metabolismo
9.
mSphere ; 5(4)2020 08 19.
Artigo em Inglês | MEDLINE | ID: mdl-32817376

RESUMO

In the Plasmodium life cycle, two infectious stages of parasites, merozoites and sporozoites, share rhoptry and microneme apical structures. A crucial step during merozoite invasion of erythrocytes is the discharge to the host cell membrane of some rhoptry neck proteins as a complex, followed by the formation of a moving junction involving the parasite-secreted protein AMA1 on the parasite membrane. Components of the merozoite rhoptry neck protein complex are also expressed in sporozoites, namely, RON2, RON4, and RON5, suggesting that invasion mechanism elements might be conserved between these infective stages. Recently, we demonstrated that RON2 is required for sporozoite invasion of mosquito salivary gland cells and mammalian hepatocytes, using a sporozoite stage-specific gene knockdown strategy in the rodent malaria parasite model, Plasmodium berghei Here, we use a coimmunoprecipitation assay and oocyst-derived sporozoite extracts to demonstrate that RON2, RON4, and RON5 also form a complex in sporozoites. The sporozoite stage-specific gene knockdown strategy revealed that both RON4 and RON5 have crucial roles during sporozoite invasion of salivary glands, including a significantly reduced attachment ability required for the onset of gliding. Further analyses indicated that RON2 and RON4 reciprocally affect trafficking to rhoptries in developing sporozoites, while RON5 is independently transported. These findings indicate that the interaction between RON2 and RON4 contributes to their stability and trafficking to rhoptries, in addition to involvement in sporozoite attachment.IMPORTANCE Sporozoites are the motile infectious stage that mediates malaria parasite transmission from mosquitoes to the mammalian host. This study addresses the question whether the rhoptry neck protein complex forms and functions in sporozoites, in addition to its role in merozoites. By applying coimmunoprecipitation and sporozoite stage-specific gene knockdown assays, it was demonstrated that RON2, RON4, and RON5 form a complex and are involved in sporozoite invasion of salivary glands via their attachment ability. These findings shed light on the conserved invasion mechanisms among apicomplexan infective stages. In addition, the sporozoite stage-specific gene knockdown system has revealed for the first time in Plasmodium that the RON2 and RON4 interaction reciprocally affects their stability and trafficking to rhoptries. Our study raises the possibility that the RON complex functions during sporozoite maturation as well as migration toward and invasion of target cells.


Assuntos
Interações Hospedeiro-Parasita , Plasmodium berghei/química , Proteínas de Protozoários/química , Glândulas Salivares/parasitologia , Esporozoítos/fisiologia , Animais , Culicidae/parasitologia , Feminino , Camundongos , Camundongos Endogâmicos ICR , Plasmodium berghei/genética , Plasmodium berghei/fisiologia , Transporte Proteico , Proteínas de Protozoários/genética , Ratos , Esporozoítos/química
10.
Vaccine ; 38(15): 3129-3136, 2020 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-32147299

RESUMO

Malaria transmission-blocking vaccines (TBVs) aim to inhibit parasite fertilization or further development within the mosquito midgut. Because TBV-immunized individuals reduce the transmission of malaria parasites to mosquito vectors, TBVs could serve as a promising strategy to eliminate malaria. We previously reported that a male specific protein, PyMiGS (Plasmodium yoelii microgamete surface protein), is localized to the surface of microgametes and anti-PyMiGS antibodies have strong transmission-blocking activity. In this study we determine a region of PyMiGS that contains epitopes inducing potent transmission-blocking antibodies. PyMiGS excluding the N-terminal signal sequence and C-terminal hydrophobic region (PyMiGS-full) was divided into five overlapping regions, named I through V, and corresponding truncated recombinant proteins were produced. Anti-region V antibody, affinity-purified from anti-PyMiGS-full rabbit antiserum, significantly reduced the number of oocysts in a mosquito membrane-feeding assay. Antibodies from mice immunized with PyMiGS-V recognized the microgamete surface and showed higher transmission-blocking efficacy than antibodies obtained by PyMiGS-full immunization. These results indicate that the major epitopes for transmission-blocking antibodies are within region V at the C-terminal region of PyMiGS. Therefore, region V of MiGS could be a promising pre-fertilization TBV candidate antigen.


Assuntos
Antígenos de Protozoários/imunologia , Antígenos de Superfície/imunologia , Vacinas Antimaláricas/imunologia , Malária/prevenção & controle , Plasmodium yoelii , Animais , Anticorpos Antiprotozoários/imunologia , Culicidae , Epitopos/imunologia , Malária/transmissão , Masculino , Camundongos , Plasmodium yoelii/imunologia , Coelhos
11.
Mol Biochem Parasitol ; 236: 111261, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31981605

RESUMO

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.


Assuntos
Plasmodium falciparum , Plasmodium yoelii , Proteínas de Protozoários/ultraestrutura , Eritrócitos/parasitologia , Malária/parasitologia , Microscopia Imunoeletrônica/métodos , Organelas/metabolismo , Organelas/ultraestrutura , Plasmodium falciparum/metabolismo , Plasmodium falciparum/ultraestrutura , Plasmodium yoelii/metabolismo , Plasmodium yoelii/ultraestrutura , Proteínas de Protozoários/metabolismo
12.
Parasitol Int ; 75: 102003, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31669509

RESUMO

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.


Assuntos
Eritrócitos/parasitologia , Proteínas de Membrana/genética , Plasmodium falciparum/genética , Proteínas de Protozoários/genética , Vacúolos/parasitologia , Animais , Membrana Celular/parasitologia , Citoplasma/parasitologia , Proteínas de Membrana/metabolismo , Microscopia Imunoeletrônica , Plasmodium falciparum/metabolismo , Transporte Proteico , Proteínas de Protozoários/metabolismo , Coelhos
13.
Expert Rev Vaccines ; 18(10): 1017-1027, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31566026

RESUMO

Introduction: Highly effective malaria vaccines are essential component toward malaria elimination. Although the leading malaria vaccine, RTS,S/AS01, with modest efficacy is being evaluated in a pilot feasibility trial, development of a malaria transmission-blocking vaccine (TBV) could make a major contribution toward malaria elimination. Only a few TBV antigens have reached pre-clinical or clinical development but with several challenges including difficulties in the expression of malaria recombinant proteins and low immunogenicity in humans. Therefore, novel approaches to accelerate TBV research to preclinical development are critical to generate an efficacious TBV.Areas covered: PubMed was searched to review the progress and future prospects of malaria TBV research and development. We also reviewed registered trials at ClinicalTrials.gov as well as post-genome TBV candidate discovery research including our efforts.Expert opinion: Wheat germ cell-free protein synthesis technology can accelerate TBV development by overcoming some current challenges of TBV research.


Assuntos
Sistema Livre de Células , Vacinas Antimaláricas/imunologia , Malária/prevenção & controle , Bases de Dados Factuais , Células Germinativas , Humanos , Imunogenicidade da Vacina , Malária/transmissão , Malária Falciparum/prevenção & controle , Plasmodium falciparum/genética , Plasmodium falciparum/imunologia , Proteínas de Protozoários/imunologia , Proteínas Recombinantes , Triticum
14.
Artigo em Inglês | MEDLINE | ID: mdl-31552198

RESUMO

In the Plasmodium lifecycle two infectious stages of parasites, merozoites, and sporozoites, efficiently infect mammalian host cells, erythrocytes, and hepatocytes, respectively. The apical structure of merozoites and sporozoites contains rhoptry and microneme secretory organelles, which are conserved with other infective forms of apicomplexan parasites. During merozoite invasion of erythrocytes, some rhoptry proteins are secreted to form a tight junction between the parasite and target cell, while others are discharged to maintain subsequent infection inside the parasitophorous vacuole. It has been questioned whether the invasion mechanisms mediated by rhoptry proteins are also involved in sporozoite invasion of two distinct target cells, mosquito salivary glands and mammalian hepatocytes. Recently we demonstrated that rhoptry neck protein 2 (RON2), which is crucial for tight junction formation in merozoites, is also important for sporozoite invasion of both target cells. With the aim of comprehensively describing the mechanisms of sporozoite invasion, the expression and localization profiles of rhoptry proteins were investigated in Plasmodium berghei sporozoites. Of 12 genes representing merozoite rhoptry molecules, nine are transcribed in oocyst-derived sporozoites at a similar or higher level compared to those in blood-stage schizonts. Immuno-electron microscopy demonstrates that eight proteins, namely RON2, RON4, RON5, ASP/RON1, RALP1, RON3, RAP1, and RAMA, localize to rhoptries in sporozoites. It is noteworthy that most rhoptry neck proteins in merozoites are localized throughout rhoptries in sporozoites. This study demonstrates that most rhoptry proteins, except components of the high-molecular mass rhoptry protein complex, are commonly expressed in merozoites and sporozoites in Plasmodium spp., which suggests that components of the invasion mechanisms are basically conserved between infective forms independently of their target cells. Combined with sporozoite-stage specific gene silencing strategies, the contribution of rhoptry proteins in invasion mechanisms can be described.


Assuntos
Perfilação da Expressão Gênica , Merozoítos/química , Plasmodium berghei/química , Proteínas de Protozoários/análise , Esporozoítos/química , Animais , Anopheles , Western Blotting , Células Cultivadas , Células Epiteliais/parasitologia , Hepatócitos/parasitologia , Mamíferos , Merozoítos/genética , Microscopia Imunoeletrônica , Organelas/química , Plasmodium berghei/genética , Transporte Proteico , Reação em Cadeia da Polimerase em Tempo Real , Esporozoítos/genética
15.
Int J Parasitol ; 49(9): 725-735, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31247198

RESUMO

The malaria parasite sporozoite sequentially invades mosquito salivary glands and mammalian hepatocytes; and is the Plasmodium lifecycle infective form mediating parasite transmission by the mosquito vector. The identification of several sporozoite-specific secretory proteins involved in invasion has revealed that sporozoite motility and specific recognition of target cells are crucial for transmission. It has also been demonstrated that some components of the invasion machinery are conserved between erythrocytic asexual and transmission stage parasites. The application of a sporozoite stage-specific gene knockdown system in the rodent malaria parasite, Plasmodium berghei, enables us to investigate the roles of such proteins previously intractable to study due to their essentiality for asexual intraerythrocytic stage development, the stage at which transgenic parasites are derived. Here, we focused on the rhoptry neck protein 11 (RON11) that contains multiple transmembrane domains and putative calcium-binding EF-hand domains. PbRON11 is localised to rhoptry organelles in both merozoites and sporozoites. To repress PbRON11 expression exclusively in sporozoites, we produced transgenic parasites using a promoter-swapping strategy. PbRON11-repressed sporozoites showed significant reduction in attachment and motility in vitro, and consequently failed to efficiently invade salivary glands. PbRON11 was also determined to be essential for sporozoite infection of the liver, the first step during transmission to the vertebrate host. RON11 is demonstrated to be crucial for sporozoite invasion of both target host cells - mosquito salivary glands and mammalian hepatocytes - via involvement in sporozoite motility.


Assuntos
Anopheles/parasitologia , Hepatócitos/parasitologia , Plasmodium berghei/fisiologia , Proteínas de Protozoários/fisiologia , Animais , Southern Blotting , DNA de Protozoário/química , DNA de Protozoário/isolamento & purificação , Eritrócitos/parasitologia , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Microscopia Eletrônica de Transmissão , Microscopia Imunoeletrônica , Proteínas de Protozoários/imunologia , Coelhos , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Glândulas Salivares/parasitologia , Esporozoítos/fisiologia
16.
Vaccine ; 37(13): 1799-1806, 2019 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-30824357

RESUMO

A transmission-blocking vaccine (TBV) against Plasmodium falciparum is likely to be a valuable tool in a malaria eradication program. Pfs230 is one of the major TBV candidates, and multiple Pfs230-based vaccines induced antibodies, which prevented oocyst formation in mosquitoes as determined by a standard membrane-feeding assay (SMFA). Pfs230 is a >300 kDa protein consisting of 14 cysteine motif (CM) domains, and the size and cysteine-rich nature of the molecule have hampered its production as an intact protein. Except for one early study with maltose-binding protein fusion Pfs230 constructs expressed in Esherichia coli, all other studies have focused on only the first four CM domains in the Pfs230 molecule. To identify all possible TBV candidate domains, we systematically produced either single-CM-domain (a total of 14), 2-CM-domain (7), or 4-CM-domain (6) recombinant protein fragments using a eukaryotic wheat germ cell-free expression system (WGCFS). In addition, two more constructs which covered previously published regions, and an N-terminal prodomain construct spanning the natural cleavage site of Pfs230 were produced. Antisera against each fragment were generated in mice and we evaluated the reactivity to native Pfs230 protein by Western blots and immunofluorescence assay (IFA), and functionality by SMFA. All 30 WGCFS-produced Pfs230 constructs were immunogenic in mice. Approximately half of the mouse antibodies specifically recognized native Pfs230 by Western blots with variable band intensities. Among them, seven antibodies showed higher reactivities against native Pfs230 determined by IFA. Interestingly, antibodies against all protein fragments containing CM domain 1 displayed strong inhibitions in SMFA, while antibodies generated using constructs without CM domain 1 showed no inhibition. The results strongly support the concept that future Pfs230-based vaccine development should focus on the Pfs230 CM domain 1.


Assuntos
Anticorpos Bloqueadores/imunologia , Anticorpos Antiprotozoários/imunologia , Vacinas Antimaláricas/imunologia , Malária Falciparum/prevenção & controle , Plasmodium falciparum/imunologia , Animais , Antígenos de Protozoários/imunologia , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Humanos , Imunogenicidade da Vacina , Imunoglobulina G/imunologia , Vacinas Antimaláricas/administração & dosagem , Malária Falciparum/transmissão , Camundongos , Proteínas de Protozoários/imunologia , Proteínas Recombinantes/imunologia
17.
Parasitol Int ; 68(1): 17-23, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30290224

RESUMO

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.


Assuntos
Interações Hospedeiro-Parasita/genética , Plasmodium/genética , Proteínas de Protozoários/genética , Esporozoítos/genética , Animais , Proteínas de Transporte , Eritrócitos/parasitologia , Feminino , Deleção de Genes , Células Hep G2 , Hepatócitos/parasitologia , Humanos , Merozoítos/genética , Merozoítos/metabolismo , Camundongos , Organelas/metabolismo , Plasmodium/crescimento & desenvolvimento , Plasmodium/imunologia , Plasmodium/fisiologia , Plasmodium berghei/genética , Plasmodium berghei/crescimento & desenvolvimento , Plasmodium berghei/fisiologia , Proteínas de Protozoários/imunologia , Esporozoítos/crescimento & desenvolvimento , Esporozoítos/fisiologia
18.
Parasitol Int ; 69: 25-29, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30385417

RESUMO

Merozoite surface proteins (MSPs) are considered as promising blood-stage malaria vaccine candidates. MSP3 has long been evaluated for its vaccine candidacy, however, the candidacy of other members of MSP3 family is insufficiently characterized. Here, we investigated Plasmodium falciparum MSP11 (PF3D7_1036000), a member of the MSP3 family, for its potential as a blood-stage vaccine candidate. The full-length protein (MSP11-FL) as well as the N-terminal half-MSP11 (MSP11-N), known to be unique among the MSP3 family members, were expressed by wheat germ cell-free system, and used to raise antibodies in rabbit. Immunoblot analysis of schizont lysates probed with anti-MSP11-N antibodies detected double bands at approximately 40 and 60 kDa, consistent with the previous report thus confirming antibodies specificity. However, inconsistent with previously reported merozoite's surface localization, immunofluorescence assay (IFA) revealed that MSP11 likely localizes to rhoptry neck of merozoites in mature schizonts. After invasion, MSP11 localized to parasitophorous vacuole and thereafter in Maurer's clefts in trophozoites. Anti-MSP11-FL antibody levels were significantly higher in asymptomatic than symptomatic P. falciparum cases in malaria low endemic Thailand. This reconfirmed that anti-MSP11 antibodies play an important role in protection against clinical malaria, as previously reported. Furthermore, in vitro growth inhibition assay revealed that anti-MSP11-FL rabbit antibodies biologically function by inhibiting merozoite invasion of erythrocytes. These findings further support the vaccine candidacy of MSP11.


Assuntos
Anticorpos Antiprotozoários/farmacologia , Antígenos de Protozoários/imunologia , Eritrócitos/parasitologia , Imunoglobulina G/farmacologia , Merozoítos/efeitos dos fármacos , Plasmodium falciparum/efeitos dos fármacos , Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/genética , Infecções Assintomáticas , Ensaio de Imunoadsorção Enzimática , Imunofluorescência , Humanos , Malária Falciparum/imunologia , Proteínas de Protozoários/genética , Tailândia
19.
Cell Microbiol ; 21(1): e12964, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30307699

RESUMO

Malaria parasite transmission to humans is initiated by the inoculation of Plasmodium sporozoites into the skin by mosquitoes. Sporozoites develop within mosquito midgut oocysts, first invade the salivary glands of mosquitoes, and finally infect hepatocytes in mammals. The apical structure of sporozoites is conserved with the infective forms of other apicomplexan parasites that have secretory organelles, such as rhoptries and micronemes. Because some rhoptry proteins are crucial for Plasmodium merozoite infection of erythrocytes, we examined the roles of rhoptry proteins in sporozoites. Here, we demonstrate that rhoptry neck protein 2 (RON2) is also localized to rhoptries in sporozoites. To elucidate RON2 function in sporozoites, we applied a promoter swapping strategy to restrict ron2 transcription to the intraerythrocytic stage in the rodent malaria parasite, Plasmodium berghei. Ron2 knockdown sporozoites were severely impaired in their ability to invade salivary glands, via decreasing the attachment capacity to the substrate. This is the first rhoptry protein demonstrated to be involved in salivary gland invasion. In addition, ron2 knockdown sporozoites showed less infectivity to hepatocytes, possibly due to decreased attachment/gliding ability, indicating that parts of the parasite invasion machinery are conserved, but their contribution might differ among infective forms. Our sporozoite stage-specific knockdown system will help to facilitate understanding the comprehensive molecular mechanisms of parasite invasion of target cells.


Assuntos
Culicidae/parasitologia , Plasmodium berghei/crescimento & desenvolvimento , Proteínas de Protozoários/metabolismo , Glândulas Salivares/parasitologia , Esporozoítos/crescimento & desenvolvimento , Fatores de Virulência/metabolismo , Animais , Técnicas de Silenciamento de Genes , Humanos , Plasmodium berghei/metabolismo , Esporozoítos/metabolismo
20.
Artigo em Inglês | MEDLINE | ID: mdl-30547015

RESUMO

Within the liver, Plasmodium sporozoites traverse cells searching for a "suitable" hepatocyte, invading these cells through a process that results in the formation of a parasitophorous vacuole (PV), within which the parasite undergoes intracellular replication as a liver stage. It was previously established that two members of the Plasmodium s48/45 protein family, P36 and P52, are essential for productive invasion of host hepatocytes by sporozoites as their simultaneous deletion results in growth-arrested parasites that lack a PV. Recent studies point toward a pathway of entry possibly involving the interaction of P36 with hepatocyte receptors EphA2, CD81, and SR-B1. However, the relationship between P36 and P52 during sporozoite invasion remains unknown. Here we show that parasites with a single P52 or P36 gene deletion each lack a PV after hepatocyte invasion, thereby pheno-copying the lack of a PV observed for the P52/P36 dual gene deletion parasite line. This indicates that both proteins are equally important in the establishment of a PV and act in the same pathway. We created a Plasmodium yoelii P36mCherry tagged parasite line that allowed us to visualize the subcellular localization of P36 and found that it partially co-localizes with P52 in the sporozoite secretory microneme organelles. Furthermore, through co-immunoprecipitation studies in vivo, we determined that P36 and P52 form a protein complex in sporozoites, indicating a concerted function for both proteins within the PV formation pathway. However, upon sporozoite stimulation, only P36 was released as a secreted protein while P52 was not. Our results support a model in which the putatively glycosylphosphatidylinositol (GPI)-anchored P52 may serve as a scaffold to facilitate the interaction of secreted P36 with the host cell during sporozoite invasion of hepatocytes.


Assuntos
Hepatócitos/parasitologia , Malária/metabolismo , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Esporozoítos/metabolismo , Animais , Antígenos CD36/metabolismo , Culicidae , Citoplasma/metabolismo , Feminino , Deleção de Genes , Técnicas de Inativação de Genes , Glicosilfosfatidilinositóis , Hepatócitos/patologia , Camundongos , Camundongos Endogâmicos BALB C , Plasmodium yoelii/metabolismo , Receptor EphA2/metabolismo , Glândulas Salivares/parasitologia , Glândulas Salivares/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...