Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
1.
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
2.
Cell Microbiol ; 20(5): e12821, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29316140

RESUMO

Anopheles mosquitoes transmit Plasmodium parasites of mammals, including the species that cause malaria in humans. Malaria pathology is caused by rapid multiplication of parasites in asexual intraerythrocytic cycles. Sexual stage parasites are also produced during the intraerythrocytic cycle and are ingested by the mosquito, initiating gametogenesis and subsequent sporogonic stage development. Here, we present a Plasmodium protein, termed microgamete surface protein (MiGS), which has an important role in male gametocyte osmiophilic body (MOB) formation and microgamete function. MiGS is expressed exclusively in male gametocytes and microgametes, in which MiGS localises to the MOB and microgamete surface. Targeted gene disruption of MiGS in a rodent malaria parasite Plasmodium yoelii 17XNL generated knockout parasites (ΔPyMiGS) that proliferate normally in erythrocytes and form male and female gametocytes. The number of MOB in male gametocyte cytoplasm is markedly reduced and the exflagellation of microgametes is impaired in ΔPyMiGS. In addition, anti-PyMiGS antibody severely blocked the parasite development in the Anopheles stephensi mosquito. MiGS might thus be a potential novel transmission-blocking vaccine target candidate.


Assuntos
Gametogênese/genética , Células Germinativas/crescimento & desenvolvimento , Malária/genética , Plasmodium yoelii/genética , Animais , Eritrócitos/parasitologia , Feminino , Células Germinativas/metabolismo , Humanos , Malária/parasitologia , Masculino , Proteínas de Membrana/genética , Plasmodium yoelii/patogenicidade , Roedores/parasitologia
3.
Malar J ; 17(1): 466, 2018 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-30545367

RESUMO

BACKGROUND: For the success of the malaria control and eradication programme it is essential to reduce parasite transmission by mosquito vectors. In the midguts of mosquitoes fed with parasite-infected blood, sexual-stage parasites fertilize to develop into motile ookinetes that traverse midgut epithelial cells and reside adjacent the basal lamina. Therefore, the ookinete is a promising target of transmission-blocking vaccines to break the parasite lifecycle in mosquito vectors. However, the molecular mechanisms of ookinete formation and invasion of epithelial cells have not been fully elucidated. A unique structure called the crystalloid body has been identified in the ookinete cytoplasm by electron microscopy, but its biological functions remain unclear. METHODS: A recombinant protein of a novel molecule, designated as crystalloid body specific PH domain-containing protein of Plasmodium yoelii (PyCryPH), was synthesized using a wheat germ cell-free system. Specific rabbit antibodies against PyCryPH were obtained to characterize the expression and localization of PyCryPH during sexual-stage parasite development. In addition, PyCryPH knockout parasites were generated by targeted gene disruption to examine PyCryPH function in mosquito-stage parasite development. RESULTS: Western blot and immunofluorescence assays using specific antibodies showed that PyCryPH is specifically expressed in zygotes and ookinetes. By immunoelectron microscopy it was demonstrated that PyCryPH is localized within crystalloid bodies. Parasites with a disrupted PyCryPH gene developed normally into ookinetes and formed oocysts on the basal lamina of midguts. In addition, the number of sporozoites residing in salivary glands was comparable to that of wild-type parasites. CONCLUSIONS: CryPH, containing a signal peptide and PH domain, is predominantly expressed in zygotes and ookinetes and is localized to crystalloid bodies in P. yoelii. CryPH accumulates in vesicle-like structures prior to the appearance of typical crystalloid bodies. Unlike other known crystalloid body localized proteins, CryPH does not appear to have a multiple domain architecture characteristic of the LAP/CCp family proteins. Although CryPH is highly conserved among Plasmodium, Babesia, Theileria, and Cryptosporidium, PyCryPH is dispensable for the development of invasive ookinetes and sporozoites in mosquito bodies.


Assuntos
Estágios do Ciclo de Vida/fisiologia , Plasmodium yoelii/química , Domínios de Homologia à Plecstrina , Proteínas de Protozoários/química , Animais , Anticorpos Antiprotozoários , Sistema Livre de Células , Malária/parasitologia , Malária/prevenção & controle , Vacinas Antimaláricas , Plasmodium yoelii/genética , Plasmodium yoelii/imunologia , Proteínas de Protozoários/genética , Proteínas de Protozoários/imunologia , Coelhos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia
4.
J Immunol ; 191(2): 795-809, 2013 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-23776179

RESUMO

The development of effective malaria vaccines and immune biomarkers of malaria is a high priority for malaria control and elimination. Ags expressed by merozoites of Plasmodium falciparum are likely to be important targets of human immunity and are promising vaccine candidates, but very few Ags have been studied. We developed an approach to assess Ab responses to a comprehensive repertoire of merozoite proteins and investigate whether they are targets of protective Abs. We expressed 91 recombinant proteins, located on the merozoite surface or within invasion organelles, and screened them for quality and reactivity to human Abs. Subsequently, Abs to 46 proteins were studied in a longitudinal cohort of 206 Papua New Guinean children to define Ab acquisition and associations with protective immunity. Ab responses were higher among older children and those with active parasitemia. High-level Ab responses to rhoptry and microneme proteins that function in erythrocyte invasion were identified as being most strongly associated with protective immunity compared with other Ags. Additionally, Abs to new or understudied Ags were more strongly associated with protection than were Abs to current vaccine candidates that have progressed to phase 1 or 2 vaccine trials. Combinations of Ab responses were identified that were more strongly associated with protective immunity than responses to their single-Ag components. This study identifies Ags that are likely to be key targets of protective human immunity and facilitates the prioritization of Ags for further evaluation as vaccine candidates and/or for use as biomarkers of immunity in malaria surveillance and control.


Assuntos
Vacinas Antimaláricas/imunologia , Malária Falciparum/imunologia , Malária Falciparum/prevenção & controle , Merozoítos/imunologia , Plasmodium falciparum/imunologia , Adolescente , Antígenos de Protozoários/imunologia , Biomarcadores/sangue , Criança , Pré-Escolar , Feminino , Humanos , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Masculino , Parasitemia/imunologia , Proteínas de Protozoários/imunologia
5.
Infect Immun ; 82(11): 4643-53, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25156733

RESUMO

Plasmodium sporozoites develop within oocysts in the mosquito midgut wall and then migrate to the salivary glands. After transmission, they embark on a complex journey to the mammalian liver, where they infect hepatocytes. Proteins on the sporozoite surface likely mediate multiple steps of this journey, yet only a few sporozoite surface proteins have been described. Here, we characterize a novel, conserved sporozoite surface protein (SSP3) in the rodent malaria parasite Plasmodium yoelii. SSP3 is a putative type I transmembrane protein unique to Plasmodium. By using epitope tagging and SSP3-specific antibodies in conjunction with immunofluorescence microscopy, we showed that SSP3 is expressed in mosquito midgut oocyst sporozoites, exhibiting an intracellular localization. In sporozoites derived from the mosquito salivary glands, however, SSP3 localized predominantly to the sporozoite surface as determined by immunoelectron microscopy. However, the ectodomain of SSP3 appeared to be inaccessible to antibodies in nonpermeabilized salivary gland sporozoites. Antibody-induced shedding of the major surface protein circumsporozoite protein (CSP) exposed the SSP3 ectodomain to antibodies in some sporozoites. Targeted deletion of SSP3 adversely affected in vitro sporozoite gliding motility, which, surprisingly, impacted neither their cell traversal capacity, host cell invasion in vitro, nor infectivity in vivo. Together, these data reveal a previously unappreciated complexity of the Plasmodium sporozoite surface proteome and the roles of surface proteins in distinct biological activities of sporozoites.


Assuntos
Regulação da Expressão Gênica/fisiologia , Proteínas de Membrana/metabolismo , Plasmodium yoelii/metabolismo , Proteínas de Protozoários/metabolismo , Esporozoítos/metabolismo , Animais , Anticorpos Antiprotozoários , Epitopos , Feminino , Deleção de Genes , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Movimento , Plasmodium yoelii/genética , Transporte Proteico , Proteínas de Protozoários/genética
6.
Cell Microbiol ; 15(9): 1508-1526, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23421981

RESUMO

Plasmodium falciparum (Pf) blood stages express falstatin, an inhibitor of cysteine proteases (ICP), which is implicated in regulating proteolysis during red blood cell infection. Recent data using the Plasmodium berghei rodent malaria model suggested an additional role for ICP in the infection of hepatocytes by sporozoites and during liver-stage development. Here we further characterize the role of ICP in vivo during infection with Plasmodium yoelii (Py) and Pf. We found that Py-ICP was refractory to targeted gene deletion indicating an essential function during asexual blood-stage replication, but significant downregulation of ICP using a regulated system did not impact blood-stage growth. Py-ICP localized to vesicles within the asexual blood-stage parasite cytoplasm, as well as the parasitophorous vacuole, and was exported to dynamic exomembrane structures in the infected RBC. In sporozoites, expression was observed in rhoptries, in addition to intracellular vesicles distinct from TRAP containing micronemes. During liver-stage development, Py-ICP was confined to the parasite compartment until the final phase of liver-stage development when, after parasitophorous vacuolemembrane breakdown, it was released into the infected hepatocyte. Finally, we identified the cysteine protease yoelipain-2 as a binding partner of Py-ICP during blood-stage infection. These data show that ICP may be important in regulating proteolytic processes during blood-stage development, and is likely playing a role in liver stage-hepatocyte interactions at the time of exoerythrocytic merozoite release.


Assuntos
Inibidores de Cisteína Proteinase/metabolismo , Eritrócitos/parasitologia , Membranas Intracelulares/metabolismo , Plasmodium yoelii/crescimento & desenvolvimento , Plasmodium yoelii/metabolismo , Sequência de Aminoácidos , Animais , Cisteína Proteases/metabolismo , Camundongos , Dados de Sequência Molecular , Ligação Proteica , Alinhamento de Sequência , Vacúolos/química , Vacúolos/parasitologia
7.
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
8.
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
9.
J Biol Chem ; 287(11): 7871-84, 2012 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-22253438

RESUMO

To survive within its host erythrocyte, Plasmodium falciparum must export hundreds of proteins across both its parasite plasma membrane and surrounding parasitophorous vacuole membrane, most of which are likely to use a protein complex known as PTEX (Plasmodium translocon of exported proteins). PTEX is a putative protein trafficking machinery responsible for the export of hundreds of proteins across the parasitophorous vacuole membrane and into the human host cell. Five proteins are known to comprise the PTEX complex, and in this study, three of the major stoichiometric components are investigated including HSP101 (a AAA(+) ATPase), a protein of no known function termed PTEX150, and the apparent membrane component EXP2. We show that these proteins are synthesized in the preceding schizont stage (PTEX150 and HSP101) or even earlier in the life cycle (EXP2), and before invasion these components reside within the dense granules of invasive merozoites. From these apical organelles, the protein complex is released into the host cell where it resides with little turnover in the parasitophorous vacuole membrane for most of the remainder of the following cell cycle. At this membrane, PTEX is arranged in a stable macromolecular complex of >1230 kDa that includes an ∼600-kDa apparently homo-oligomeric complex of EXP2 that can be separated from the remainder of the PTEX complex using non-ionic detergents. Two different biochemical methods undertaken here suggest that PTEX components associate as EXP2-PTEX150-HSP101, with EXP2 associating with the vacuolar membrane. Collectively, these data support the hypothesis that EXP2 oligomerizes and potentially forms the putative membrane-spanning pore to which the remainder of the PTEX complex is attached.


Assuntos
Membranas Intracelulares/metabolismo , Proteínas de Membrana/biossíntese , Complexos Multiproteicos/biossíntese , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/biossíntese , Vacúolos/metabolismo , Humanos , Proteínas de Membrana/genética , Complexos Multiproteicos/genética , Plasmodium falciparum/genética , Transporte Proteico/fisiologia , Proteínas de Protozoários/genética , Esquizontes/metabolismo , Vacúolos/genética
10.
Infect Immun ; 81(11): 4290-8, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24002067

RESUMO

Erythrocyte invasion by merozoites is an obligatory stage of Plasmodium infection and is essential to disease progression. Proteins in the apical organelles of merozoites mediate the invasion of erythrocytes and are potential malaria vaccine candidates. Rhoptry-associated, leucine zipper-like protein 1 (RALP1) of Plasmodium falciparum was previously found to be specifically expressed in schizont stages and localized to the rhoptries of merozoites by immunofluorescence assay (IFA). Also, RALP1 has been refractory to gene knockout attempts, suggesting that it is essential for blood-stage parasite survival. These characteristics suggest that RALP1 can be a potential blood-stage vaccine candidate antigen, and here we assessed its potential in this regard. Antibodies were raised against recombinant RALP1 proteins synthesized by using the wheat germ cell-free system. Immunoelectron microscopy demonstrated for the first time that RALP1 is a rhoptry neck protein of merozoites. Moreover, our IFA data showed that RALP1 translocates from the rhoptry neck to the moving junction during merozoite invasion. Growth and invasion inhibition assays revealed that anti-RALP1 antibodies inhibit the invasion of erythrocytes by merozoites. The findings that RALP1 possesses an erythrocyte-binding epitope in the C-terminal region and that anti-RALP1 antibodies disrupt tight-junction formation, are evidence that RALP1 plays an important role during merozoite invasion of erythrocytes. In addition, human sera collected from areas in Thailand and Mali where malaria is endemic recognized this protein. Overall, our findings indicate that RALP1 is a rhoptry neck erythrocyte-binding protein and that it qualifies as a potential blood-stage vaccine candidate.


Assuntos
Antígenos de Protozoários/metabolismo , Eritrócitos/parasitologia , Vacinas Antimaláricas/imunologia , Plasmodium falciparum/fisiologia , Proteínas de Protozoários/metabolismo , Receptores de Superfície Celular/metabolismo , Adulto , Animais , Anticorpos Antiprotozoários/sangue , Antígenos de Protozoários/imunologia , Imunofluorescência , Humanos , Vacinas Antimaláricas/genética , Mali , Microscopia Imunoeletrônica , Proteínas de Protozoários/imunologia , Receptores de Superfície Celular/imunologia , Soro/imunologia , Tailândia
11.
Malar J ; 12: 129, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23587146

RESUMO

BACKGROUND: Despite the development of malaria control programs, billions of people are still at risk for this infectious disease. Recently, the idea of the transmission-blocking vaccine, which works by interrupting the infection of mosquitoes by parasites, has gained attention as a promising strategy for malaria control and eradication. To date, a limited number of surface proteins have been identified in mosquito-stage parasites and investigated as potential targets for transmission-blocking vaccines. Therefore, for the development of effective transmission-blocking strategies in epidemic areas, it is necessary to identify novel zygote/ookinete surface proteins as candidate antigens. METHODS: Since the expression of many zygote/ookinete proteins is regulated post-transcriptionally, proteins that are regulated by well-known translational mediators were focused. Through in silico screening, CPW-WPC family proteins were selected as potential zygote/ookinete surface proteins. All experiments were performed in the rodent malaria parasite, Plasmodium yoelii XNL. mRNA and protein expression profiles were examined by RT-PCR and western blotting, respectively, over the course of the life cycle of the malaria parasite. Protein function was also investigated by the generation of gene-disrupted transgenic parasites. RESULTS: The CPW-WPC protein family, named after the unique WxC repeat domains, is highly conserved among Plasmodium species. It is revealed that CPW-WPC mRNA transcripts are transcribed in gametocytes, while CPW-WPC proteins are expressed in zygote/ookinete-stage parasites. Localization analysis reveals that one of the CPW-WPC family members, designated as PyCPW-WPC-1, is a novel zygote/ookinete stage-specific surface protein. Targeted disruption of the pycpw-wpc-1 gene caused no obvious defects during ookinete and oocyst formation, suggesting that PyCPW-WPC-1 is not essential for mosquito-stage parasite development. CONCLUSIONS: It is demonstrated that PyCPW-WPC-1 can be classified as a novel, post-transcriptionally regulated zygote/ookinete surface protein. Additional studies are required to determine whether all CPW-WPC family members are also present on the ookinete surface and share similar biological roles during mosquito-stage parasite development. Further investigations of CPW-WPC family proteins may facilitate understanding of parasite biology in the mosquito stage and development of transmission-blocking vaccines.


Assuntos
Antígenos de Protozoários/análise , Expressão Gênica , Proteínas de Membrana/análise , Plasmodium yoelii/química , Zigoto/química , Animais , Antígenos de Protozoários/genética , Western Blotting , Feminino , Perfilação da Expressão Gênica , Técnicas de Inativação de Genes , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Plasmodium yoelii/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA
12.
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
13.
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
14.
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
15.
Proc Natl Acad Sci U S A ; 106(17): 7167-72, 2009 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-19346470

RESUMO

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.


Assuntos
Aminoácidos/metabolismo , Eritrócitos/metabolismo , Plasmodium yoelii/metabolismo , Plasmodium yoelii/patogenicidade , Proteínas de Protozoários/metabolismo , Sequência de Aminoácidos , Aminoácidos/genética , Animais , Ligantes , Camundongos , Microscopia Imunoeletrônica , Dados de Sequência Molecular , Plasmodium yoelii/genética , Proteínas de Protozoários/química , Proteínas de Protozoários/genética , Virulência
16.
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
17.
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
18.
Infect Immun ; 79(10): 4260-75, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21807905

RESUMO

The creation of subunit vaccines to prevent malaria infection has been hampered by the intrinsically weak immunogenicity of the recombinant antigens. We have developed a novel strategy to increase immune responses by creating genetic fusion proteins to target specific antigen-presenting cells (APCs). The fusion complex was composed of three physically linked molecular entities: (i) a vaccine antigen, (ii) a multimeric α-helical coiled-coil core, and (iii) an APC-targeting ligand linked to the core via a flexible linker. The vaccine efficacy of the tricomponent complex was evaluated using an ookinete surface protein of Plasmodium vivax, Pvs25, and merozoite surface protein-1 of Plasmodium yoelii. Immunization of mice with the tricomponent complex induced a robust antibody response and conferred substantial levels of P. vivax transmission blockade as evaluated by a membrane feed assay, as well as protection from lethal P. yoelii infection. The observed effect was strongly dependent on the presence of all three components physically integrated as a fusion complex. This system, designated the tricomponent immunopotentiating system (TIPS), onto which any recombinant protein antigens or nonproteinaceous substances could be loaded, may be a promising strategy for devising subunit vaccines or adjuvants against various infectious diseases, including malaria.


Assuntos
Desenho de Fármacos , Vacinas Antimaláricas/administração & dosagem , Malária/prevenção & controle , Plasmodium vivax/imunologia , Plasmodium yoelii/imunologia , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas Sintéticas/administração & dosagem , Sequência de Aminoácidos , Animais , Células Apresentadoras de Antígenos/imunologia , Antígenos de Protozoários/química , Antígenos de Protozoários/genética , Antígenos de Protozoários/imunologia , Antígenos de Superfície/química , Antígenos de Superfície/genética , Antígenos de Superfície/imunologia , Linfócitos B/imunologia , Sequência de Bases , Proteínas da Matriz Extracelular/química , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/imunologia , Feminino , Glicoproteínas/química , Glicoproteínas/genética , Glicoproteínas/imunologia , Ligantes , Ativação Linfocitária , Malária/imunologia , Vacinas Antimaláricas/química , Vacinas Antimaláricas/genética , Vacinas Antimaláricas/imunologia , Malária Vivax/imunologia , Malária Vivax/prevenção & controle , Proteínas Matrilinas , Proteína 1 de Superfície de Merozoito/química , Proteína 1 de Superfície de Merozoito/genética , Proteína 1 de Superfície de Merozoito/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Plasmodium vivax/genética , Plasmodium yoelii/genética , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/imunologia , Vacinas de Subunidades Antigênicas/química , Vacinas de Subunidades Antigênicas/genética , Vacinas de Subunidades Antigênicas/imunologia , Vacinas Sintéticas/química , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia
19.
Infect Immun ; 79(11): 4523-32, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21896773

RESUMO

One of the solutions for reducing the global mortality and morbidity due to malaria is multivalent vaccines comprising antigens of several life cycle stages of the malarial parasite. Hence, there is a need for supplementing the current set of malaria vaccine candidate antigens. Here, we aimed to characterize glycosylphosphatidylinositol (GPI)-anchored micronemal antigen (GAMA) encoded by the PF08_0008 gene in Plasmodium falciparum. Antibodies were raised against recombinant GAMA synthesized by using a wheat germ cell-free system. Immunoelectron microscopy demonstrated for the first time that GAMA is a microneme protein of the merozoite. Erythrocyte binding assays revealed that GAMA possesses an erythrocyte binding epitope in the C-terminal region and it binds a nonsialylated protein receptor on human erythrocytes. Growth inhibition assays revealed that anti-GAMA antibodies can inhibit P. falciparum invasion in a dose-dependent manner and GAMA plays a role in the sialic acid (SA)-independent invasion pathway. Anti-GAMA antibodies in combination with anti-erythrocyte binding antigen 175 exhibited a significantly higher level of invasion inhibition, supporting the rationale that targeting of both SA-dependent and SA-independent ligands/pathways is better than targeting either of them alone. Human sera collected from areas of malaria endemicity in Mali and Thailand recognized GAMA. Since GAMA in P. falciparum is refractory to gene knockout attempts, it is essential to parasite invasion. Overall, our study indicates that GAMA is a novel blood-stage vaccine candidate antigen.


Assuntos
Antígenos de Protozoários/imunologia , Vacinas Antimaláricas/imunologia , Merozoítos/metabolismo , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/imunologia , Animais , Anticorpos Antiprotozoários , Sistema Livre de Células , Eritrócitos/citologia , Eritrócitos/metabolismo , Regulação da Expressão Gênica/fisiologia , Proteínas de Membrana/metabolismo , Microscopia Imunoeletrônica , Neuraminidase , Plasmodium falciparum/genética , Ligação Proteica , Transporte Proteico , Proteínas de Protozoários/genética
20.
Southeast Asian J Trop Med Public Health ; 42(6): 1313-21, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22299398

RESUMO

Plasmodium vivax subtelomeric transmembrane protein (PvSTP) is a homolog of P. falciparum SURFIN4.2', a protein exposed on the parasite-infected erythrocyte (iE) surface, and is thus considered to be exposed on P. vivax-iE. Because antibodies targeting antigens located on the surface of P. falciparum-iE, such as P. falciparum erythrocyte membrane protein 1, play an important role in regulating the course of disease, we evaluated the presence of antibodies in P. vivax-infected patients against two PvSTP paralogs, PvSTP1 and PvSTP2. Recombinant proteins corresponding to cysteine-rich domain (CRD) of the PvSTP extracellular region and the cytoplasmic region (CYT) were generated and used for the enzyme-linked immunosorbent assay. Plasma samples (n = 70) reacted positively with recombinant PvSTP1-CRD (40%), PvSTP1-CYT (31%), PvSTP2-CRD (27%), and PvSTP2-CYT (56%), suggesting that PvSTP1 and -2 are naturally immunogenic. Specific response against either PvSTP1 or PvSTP2 indicates the existence of specific antibodies for either PvSTP1 or -2.


Assuntos
Imunidade Humoral , Malária Vivax/imunologia , Proteínas de Membrana/imunologia , Plasmodium vivax/imunologia , Proteínas de Protozoários/imunologia , Adolescente , Adulto , Anticorpos Antiprotozoários/imunologia , Antimaláricos/uso terapêutico , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Malária Vivax/tratamento farmacológico , Masculino , Pessoa de Meia-Idade , Plasmodium falciparum/imunologia , Estatísticas não Paramétricas , Tailândia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA