Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
1.
Annu Rev Microbiol ; 76: 619-640, 2022 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-35671531

RESUMO

Apicomplexa are obligatory intracellular parasites that sense and actively invade host cells. Invasion is a conserved process that relies on the timely and spatially controlled exocytosis of unique specialized secretory organelles termed micronemes and rhoptries. Microneme exocytosis starts first and likely controls the intricate mechanism of rhoptry secretion. To assemble the invasion machinery, micronemal proteins-associated with the surface of the parasite-interact and form complexes with rhoptry proteins, which in turn are targeted into the host cell. This review covers the molecular advances regarding microneme and rhoptry exocytosis and focuses on how the proteins discharged from these two compartments work in synergy to drive a successful invasion event. Particular emphasis is given to the structure and molecular components of the rhoptry secretion apparatus, and to the current conceptual framework of rhoptry exocytosis that may constitute an unconventional eukaryotic secretory machinery closely related to the one described in ciliates.


Assuntos
Parasitos , Toxoplasma , Animais , Interações Hospedeiro-Parasita , Organelas/metabolismo , Parasitos/metabolismo , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Toxoplasma/metabolismo
2.
Mol Microbiol ; 121(3): 394-412, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-37314965

RESUMO

Plasmodium parasites, the eukaryotic pathogens that cause malaria, feature three distinct invasive forms tailored to the host environment they must navigate and invade for life cycle progression. One conserved feature of these invasive forms is the micronemes, apically oriented secretory organelles involved in egress, motility, adhesion, and invasion. Here we investigate the role of GPI-anchored micronemal antigen (GAMA), which shows a micronemal localization in all zoite forms of the rodent-infecting species Plasmodium berghei. ∆GAMA parasites are severely defective for invasion of the mosquito midgut. Once formed, oocysts develop normally, however, sporozoites are unable to egress and exhibit defective motility. Epitope-tagging of GAMA revealed tight temporal expression late during sporogony and showed that GAMA is shed during sporozoite gliding motility in a similar manner to circumsporozoite protein. Complementation of P. berghei knockout parasites with full-length P. falciparum GAMA partially restored infectivity to mosquitoes, indicating conservation of function across Plasmodium species. A suite of parasites with GAMA expressed under the promoters of CTRP, CAP380, and TRAP, further confirmed the involvement of GAMA in midgut infection, motility, and vertebrate infection. These data show GAMA's involvement in sporozoite motility, egress, and invasion, implicating GAMA as a regulator of microneme function.


Assuntos
Culicidae , Parasitos , Animais , Culicidae/metabolismo , Culicidae/parasitologia , Parasitos/metabolismo , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Oocistos , Plasmodium berghei/genética , Plasmodium berghei/metabolismo , Esporozoítos/metabolismo
3.
Infect Immun ; 92(2): e0045623, 2024 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-38179959

RESUMO

Using transgenic Eimeria spp. to deliver exogenous antigens is a viable option for developing multivalent live vaccines. Previous research revealed that the location of antigen expression in recombinant Eimeria dictates the magnitude and type of immune responses. In this study, we constructed genetically modified Eimeria acervulina that expressed VP2 protein, a protective antigen from infectious bursal disease virus (IBDV), on the surface or in the microneme of sporozoites. After vaccination, VP2-specific antibody was readily detected in specific pathogen-free chickens receiving transgenic E. acervulina parasites expressing VP2 in microneme, but animals vaccinated with which expressing VP2 on surface failed to produce detectable antibody after two times immunizations. Moreover, the bursal lesion of microneme-located VP2 transgenic E. acervulina immunized chickens was less severe compared with un-immunized animals after IBDV challenge infection. Therefore, genetically modified E. acervulina that express IBDV-derived VP2 in micronemes are effective in inducing specific antibody responses against VP2, while parasites that have VP2 expression on cell surface are not suitable. Thus, the use of Eimeria parasites as vaccine vectors needs to consider the proper targeting of exogenous immunogens. Our results have implications for the design of other vector vaccines.


Assuntos
Eimeria , Vírus da Doença Infecciosa da Bursa , Doenças das Aves Domésticas , Vacinas , Animais , Galinhas , Eimeria/genética , Vírus da Doença Infecciosa da Bursa/metabolismo , Micronema , Doenças das Aves Domésticas/prevenção & controle , Anticorpos Antivirais/metabolismo
4.
Annu Rev Microbiol ; 73: 579-599, 2019 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-31500539

RESUMO

The Apicomplexa phylum includes a large group of obligate intracellular protozoan parasites responsible for important diseases in humans and animals. Toxoplasma gondii is a widespread parasite with considerable versatility, and it is capable of infecting virtually any warm-blooded animal, including humans. This outstanding success can be attributed at least in part to an efficient and continuous sensing of the environment, with a ready-to-adapt strategy. This review updates the current understanding of the signals governing the lytic cycle of T. gondii, with particular focus on egress from infected cells, a key step for balancing survival, multiplication, and spreading in the host. We cover the recent advances in the conceptual framework of regulation of microneme exocytosis that ensures egress, motility, and invasion. Particular emphasis is given to the trigger molecules and signaling cascades regulating exit from host cells.


Assuntos
Secreções Corporais/parasitologia , Interações Hospedeiro-Parasita/fisiologia , Transdução de Sinais , Toxoplasma , Actomiosina , Animais , Secreções Corporais/metabolismo , Sinalização do Cálcio , Adesão Celular , Movimento Celular , Humanos , Proteínas Motores Moleculares/metabolismo , Organelas/metabolismo , Proteínas de Protozoários/metabolismo , Toxoplasma/metabolismo , Toxoplasma/ultraestrutura
5.
Parasitol Res ; 123(2): 139, 2024 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-38381180

RESUMO

The flow of calcium ions (Ca2+) is involved in numerous vital activities of Toxoplasma gondii. Calreticulin is a type of Ca2+-binding protein in the endoplasmic reticulum (ER) that is involved in Ca2+ signaling pathway regulation, Ca2+ storage, and protein folding. In this work, the calreticulin (CALR), a protein predicted to possess a conserved domain of calreticulin in T. gondii, was characterized. The CALR localized in the ER. Using reverse genetics, we discovered that CALR is not necessary for the lytic cycle, including invasion and replication. However, depletion of CALR affected microneme secretion triggered by A23187, which is a Ca2+ ionophore used to increase cytoplasmic Ca2+ concentration. Furthermore, we discovered that CALR influences Ca2+ release. Transcriptomic comparison between Δcalr and Δku80 parasites showed that 226 genes in the Δcalr parasites were significantly downregulated (p < 0.05). The cellular biological functions of the downregulated genes were mainly involved in calmodulin-dependent protein kinase pathways. Furthermore, in the absence of CALR, tachyzoites were still able to cause acute infection in mice. These results imply that by influencing ER Ca2+ release content, CALR may further impair the ionophore-induced secretion of the parasite. However, this protein is not required for the completion of the parasite's lytic cycle or for the acute virulence of the parasite.


Assuntos
Calreticulina , Proteínas de Protozoários , Toxoplasma , Animais , Camundongos , Calreticulina/genética , Calreticulina/metabolismo , Retículo Endoplasmático , Ionóforos , Micronema , Toxoplasma/genética , Toxoplasma/metabolismo , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo
6.
J Biol Chem ; 298(4): 101765, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35202655

RESUMO

Glycosylphosphatidylinositol-anchored micronemal antigen (GAMA) is an erythrocyte binding protein known to be involved in malarial parasite invasion. Although anti-GAMA antibodies have been shown to block GAMA attachment to the erythrocyte surface and subsequently inhibit parasite invasion, little is known about the molecular mechanisms by which GAMA promotes the invasion process. In this study, LC-MS analysis was performed on the erythrocyte membrane to identify the specific receptor that interacts with GAMA. We found that ankyrin 1 and the band 3 membrane protein showed affinity for GAMA, and characterization of their binding specificity indicated that both Plasmodium falciparum and Plasmodium vivax GAMA bound to the same extracellular loop of band 3 (loop 5). In addition, we show the interaction between GAMA and band 3 was sensitive to chymotrypsin. Furthermore, antibodies against band 3 loop 5 were able to reduce the binding activity of GAMA to erythrocytes and inhibit the invasion of P. falciparum merozoites into human erythrocytes, whereas antibodies against P. falciparum GAMA (PfGAMA)-Tr3 only slightly reduced P. falciparum invasion. The identification and characterization of the erythrocyte GAMA receptor is a novel finding that identifies an essential mechanism of parasite invasion of host erythrocytes.


Assuntos
Eritrócitos , Malária Falciparum , Plasmodium falciparum , Proteínas de Protozoários , Animais , Proteína 1 de Troca de Ânion do Eritrócito/metabolismo , Anquirinas/metabolismo , Eritrócitos/parasitologia , Humanos , Malária Falciparum/metabolismo , Plasmodium falciparum/metabolismo , Plasmodium vivax/metabolismo , Proteínas de Protozoários/metabolismo
7.
BMC Immunol ; 23(1): 21, 2022 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-35468726

RESUMO

BACKGROUND: Apical membrane antigen 1 (AMA1) and microneme-associated antigen (MIC) of Plasmodium parasites are important factors involved in host cell invasion. METHODS: In this study, influenza VLP vaccines containing both codon-optimized AMA1 and MIC were generated and the vaccine efficacy was evaluated in mice. RESULTS: VLPs vaccine immunization elicited higher levels of parasite-specific IgG and IgG2a antibody responses in sera. CD4+ and CD8+ T cells and germinal center B cells in blood, inguinal lymph nodes (ILN) and spleen were found to be significantly increased. Importantly, VLPs vaccination significantly reduced the levels of pro-inflammatory cytokines IFN-γ and TNF-α, decreased parasitemia in blood, resulting in lower body weight loss and longer survival time compared to control. CONCLUSION: These results indicated that VLPs containing P. berghei AMA1 and MIC could be a candidate for malaria blood-stage vaccine design.


Assuntos
Influenza Humana , Vacinas Antimaláricas , Orthomyxoviridae , Vacinas de Partículas Semelhantes a Vírus , Animais , Linfócitos T CD8-Positivos , Humanos , Camundongos , Micronema , Plasmodium berghei , Proteínas de Protozoários
8.
Traffic ; 20(8): 583-600, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31206964

RESUMO

To efficiently enter host cells, apicomplexan parasites such as Toxoplasma gondii rely on an apical complex composed of tubulin-based structures as well as two sets of secretory organelles named micronemes and rhoptries. The trafficking and docking of these organelles to the apical pole of the parasite is crucial for the discharge of their contents. Here, we describe two proteins typically associated with microtubules, Centrin 2 (CEN2) and Dynein Light Chain 8a (DLC8a), that are required for efficient host cell invasion. CEN2 localizes to four different compartments, and remarkably, conditional depletion of the protein occurs in stepwise manner, sequentially depleting the protein pools from each location. This phenomenon allowed us to discern the essential function of the apical pool of CEN2 for microneme secretion, motility, invasion and egress. DLC8a localizes to the conoid, and its depletion also perturbs microneme exocytosis in addition to the apical docking of the rhoptry organelles, causing a severe defect in host cell invasion. Phenotypic characterization of CEN2 and DLC8a indicates that while both proteins participate in microneme secretion, they likely act at different steps along the cascade of events leading to organelle exocytosis.


Assuntos
Dineínas/metabolismo , Exocitose , Proteínas de Protozoários/metabolismo , Toxoplasma/metabolismo , Combinação Trimetoprima e Sulfametoxazol/metabolismo , Dineínas/química , Transporte Proteico , Proteínas de Protozoários/química , Vesículas Secretórias/metabolismo , Combinação Trimetoprima e Sulfametoxazol/química
9.
EMBO Rep ; 20(6)2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31097469

RESUMO

Invasion of human red blood cells by the malaria parasite Plasmodium falciparum is an essential step in the development of the disease. Consequently, the molecular players involved in host cell invasion represent important targets for inhibitor design and vaccine development. The process of merozoite invasion is a succession of steps underlined by the sequential secretion of the organelles of the apical complex. However, little is known with regard to how their contents are exocytosed. Here, we identify a phosphoinositide-binding protein conserved in apicomplexan parasites and show that it is important for the attachment and subsequent invasion of the erythrocyte by the merozoite. Critically, removing the protein from its site of action by knock sideways preferentially prevents the secretion of certain types of micronemes. Our results therefore provide evidence for a role of phosphoinositide lipids in the malaria invasion process and provide further insight into the secretion of microneme organelle populations, which is potentially applicable to diverse apicomplexan parasites.


Assuntos
Exocitose , Plasmodium falciparum/fisiologia , Proteínas de Protozoários/metabolismo , Sequência de Aminoácidos , Sequência Conservada , Eritrócitos/parasitologia , Humanos , Estágios do Ciclo de Vida , Fosfatidilinositóis/metabolismo , Domínios de Homologia à Plecstrina , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Proteínas de Protozoários/química , Proteínas de Protozoários/genética
10.
J Eukaryot Microbiol ; 68(2): e12836, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33289220

RESUMO

Eimeria tenella is an obligate intracellular parasite in the phylum Apicomplexa. As described for other members of Apicomplexa, apical membrane antigen 1 (AMA1) has been shown to be critical for sporozoite invasion of host cells by E. tenella. Recently, an E. tenella paralogue of AMA1 (EtAMA1), dubbed sporoAMA1 (EtAMA3), was identified in proteomic and transcriptomic analyses of E. tenella, but not further characterized. Here, we show that EtAMA3 is a type I integral membrane protein that has 24% -38% identity with other EtAMAs. EtAMA3 has the same pattern of Cys residues in domains I and II of AMA1 orthologs from apicomplexan parasites, but high variance in domain III, with all six invariant Cys residues absent. EtAMA3 expression was developmentally regulated at the mRNA and protein levels. EtAMA3 protein was detected in sporulated oocysts and sporozoites, but not in the unsporulated oocysts or second-generation merozoites. EtAMA3 is secreted by micronemes and is primarily localized to the apical end of sporozoites during host-cell invasion. Additionally, pretreatment of sporozoites with rEtAMA3-specific antibodies substantially impeded their invasion into host cells. These results suggest EtAMA3 is a sporozoite-specific protein that is involved in host-cell sporozoite invasion.


Assuntos
Eimeria tenella , Animais , Eimeria tenella/genética , Merozoítos , Proteômica , Proteínas de Protozoários/genética , Esporozoítos
11.
Traffic ; 19(5): 336-353, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29437275

RESUMO

Host cytosolic proteins are endocytosed by Toxoplasma gondii and degraded in its lysosome-like compartment, the vacuolar compartment (VAC), but the dynamics and route of endocytic trafficking remain undefined. Conserved endocytic components and plant-like features suggest T. gondii endocytic trafficking involves transit through early and late endosome-like compartments (ELCs) and potentially the trans-Golgi network (TGN) as in plants. However, exocytic trafficking to regulated secretory organelles, micronemes and rhoptries, also proceeds through ELCs and requires classical endocytic components, including a dynamin-related protein, DrpB. Here, we show that host cytosolic proteins are endocytosed within 7 minutes post-invasion, trafficked through ELCs en route to the VAC, and degraded within 30 minutes. We could not definitively interpret if ingested protein is trafficked through the TGN. We also found that parasites ingest material from the host cytosol throughout the parasite cell cycle. Ingested host proteins colocalize with immature microneme proteins, proM2AP and proMIC5, in transit to the micronemes, but not with the immature rhoptry protein proRON4, indicating that endocytic trafficking of ingested protein intersects with exocytic trafficking of microneme proteins. Finally, we show that conditional expression of a DrpB dominant negative mutant increases T. gondii ingestion of host-derived proteins, suggesting that DrpB is not required for parasite endocytosis.


Assuntos
Endocitose , Exocitose , Proteínas de Protozoários/metabolismo , Toxoplasma/metabolismo , Animais , Células CHO , Cricetinae , Cricetulus , Endossomos/metabolismo , Complexo de Golgi/metabolismo
12.
Cell Microbiol ; 21(5): e13018, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30791192

RESUMO

One of the hallmarks of the parasitic phylum of Apicomplexa is the presence of highly specialised, apical secretory organelles, called the micronemes and rhoptries that play critical roles in ensuring survival and dissemination. Upon exocytosis, the micronemes release adhesin complexes, perforins, and proteases that are crucially implicated in egress from infected cells, gliding motility, migration across biological barriers, and host cell invasion. Recent studies on Toxoplasma gondii and Plasmodium species have shed more light on the signalling events and the machinery that trigger microneme secretion. Intracellular cyclic nucleotides, calcium level, and phosphatidic acid act as key mediators of microneme exocytosis, and several downstream effectors have been identified. Here, we review the key steps of microneme biogenesis and exocytosis, summarising the still fractal knowledge at the molecular level regarding the fusion event with the parasite plasma membrane.


Assuntos
Organelas/metabolismo , Toxoplasma/metabolismo , Animais , Membrana Celular/metabolismo , Membrana Celular/ultraestrutura , Exocitose/genética , Interações Hospedeiro-Parasita , Humanos , Fusão de Membrana/genética , Biogênese de Organelas , Organelas/ultraestrutura , Peptídeo Hidrolases/metabolismo , Perforina/genética , Perforina/metabolismo , Transdução de Sinais , Toxoplasma/patogenicidade , Toxoplasma/ultraestrutura
13.
Eur J Clin Microbiol Infect Dis ; 38(4): 617-629, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30680553

RESUMO

Toxoplasma gondii is an intracellular parasite infecting almost all warm-blooded animals. Many studies on vaccination have been performed previously, and micronemal proteins (MICs) have crucial importance in this regard. The current review aims to reveal the efficiency of MICs as target antigen, adjuvants, animal models (species/strain), T. gondii strains for challenge infection, and routes of vaccine to prevent Toxoplasma infection. A comprehensive literature search was performed on April 18, 2018, in several known databases. Studies were included when evaluating vaccines based on MIC against T. gondii compared to that of a control group. Two independent researchers done the search process, study choice, and data extraction. A total of 28 articles published were selected for further analysis. Among them, 57.03% of the studies focused on MIC3 and its epitopes. SAG1 was further used in cocktail vaccines compared to other antigens. GM-CSF and Freund's complete were the predominant adjuvants used. BALB/c mice have been introduced as a proper model for lethal challenge. Virulent T. gondii (RH) was utilized more than other strains for challenge. Among MICs, the results of vaccination with MIC1-4, MIC6, and PLP1 demonstrated significantly strong humoral and cellular immunity, increased survival time, and reduced cyst burden in the mice. This review summarizes the latest results on MIC-based vaccines and presents that the most effective vaccination procedure is the administration of the cocktail vaccines. Our survey can serve as a basis for further studies to develop more efficient novel vaccines against T. gondii for animals and humans.


Assuntos
Antígenos de Protozoários/imunologia , Proteínas de Protozoários/imunologia , Toxoplasma/imunologia , Toxoplasmose/prevenção & controle , Animais , Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/química , Modelos Animais de Doenças , Adjuvante de Freund/administração & dosagem , Fator Estimulador de Colônias de Granulócitos e Macrófagos/administração & dosagem , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Humanos , Imunidade Celular , Imunidade Humoral , Camundongos Endogâmicos BALB C , Toxoplasma/patogenicidade , Toxoplasmose/imunologia , Vacinação , Vacinas de DNA/administração & dosagem , Vacinas de DNA/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/imunologia
14.
Int J Mol Sci ; 20(20)2019 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-31658592

RESUMO

The microneme organelles of Toxoplasma gondii tachyzoites release protein complexes (MICs), including one composed of the transmembrane protein MIC6 plus MIC1 and MIC4. In this complex, carbohydrate recognition domains of MIC1 and MIC4 are exposed and interact with terminal sialic acid and galactose residues, respectively, of host cell glycans. Recently, we demonstrated that MIC1 and MIC4 binding to the N-glycans of Toll-like receptor (TLR) 2 and TLR4 on phagocytes triggers cell activation and pro-inflammatory cytokine production. Herein, we investigated the requirement for TLR2 heterodimerization and co-receptors in MIC-induced responses, as well as the signaling molecules involved. We used MICs to stimulate macrophages and HEK293T cells transfected with TLR2 and TLR1 or TLR6, both with or without the co-receptors CD14 and CD36. Then, the cell responses were analyzed, including nuclear factor-kappa B (NF-κB) activation and cytokine production, which showed that (1) only TLR2, among the studied factors, is crucial for MIC-induced cell activation; (2) TLR2 heterodimerization augments, but is not critical for, activation; (3) CD14 and CD36 enhance the response to MIC stimulus; and (4) MICs activate cells through a transforming growth factor beta-activated kinase 1 (TAK1)-, mammalian p38 mitogen-activated protein kinase (p38)-, and NF-κB-dependent pathway. Remarkably, among the studied factors, the interaction of MIC1 and MIC4 with TLR2 N-glycans is sufficient to induce cell activation, which promotes host protection against T. gondii infection.


Assuntos
Moléculas de Adesão Celular/química , Moléculas de Adesão Celular/metabolismo , Dimerização , Proteínas de Protozoários/química , Proteínas de Protozoários/metabolismo , Receptor 2 Toll-Like/química , Receptor 2 Toll-Like/metabolismo , Toxoplasma/metabolismo , Animais , Antígenos CD36/genética , Antígenos CD36/metabolismo , Citocinas/análise , Feminino , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Receptores de Lipopolissacarídeos/genética , Receptores de Lipopolissacarídeos/metabolismo , MAP Quinase Quinase Quinases/metabolismo , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Células RAW 264.7 , Transdução de Sinais , Receptor 1 Toll-Like/metabolismo , Receptor 6 Toll-Like/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
15.
Exp Parasitol ; 175: 21-27, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28130119

RESUMO

Microneme proteins play an important role in the invasion process of Apicomplexan parasites through adhesion to host cells. We discovered a new N. caninum protein, NcMIC8, which is highly identical to TgMIC8. The NcMIC8 sequence has 2049 bp and no intron in the open reading fragment. It has a molecular weight of 73.8 kDa and contains a signal peptide, a transmembrane region, a low complexity region and 10 epidermal growth factor (EGF) domains. Immuno-fluorescence assay showed that NcMIC8 is located in the microneme. NcMIC8 was secreted to culture medium under stimulation of 1% ethanol, and cleaved to form the mature body of 40 kDa before transporting to microneme or during secretion. Blocking NcMIC8 using anti-NcMIC8 serum effectively inhibited host cell invasion by tachyzoites in vitro. NcMIC8 in the form of mature body interacts with NcMIC3, and the two microneme proteins form a complex probably during transportation. NcMIC8 is a new microneme protein of N. caninum and could be an attractive target for the control of neosporosis.


Assuntos
Moléculas de Adesão Celular/fisiologia , Neospora/fisiologia , Proteínas de Protozoários/fisiologia , Animais , Anticorpos Antiprotozoários/imunologia , Western Blotting , Moléculas de Adesão Celular/química , Moléculas de Adesão Celular/imunologia , Chlorocebus aethiops , Coccidiose/parasitologia , Biologia Computacional , Feminino , Imunofluorescência , Imunoprecipitação , Camundongos , Camundongos Endogâmicos BALB C , Neospora/química , Neospora/imunologia , Neospora/ultraestrutura , Organelas/química , Organelas/fisiologia , Proteínas de Protozoários/química , Proteínas de Protozoários/imunologia , Coelhos , Células Vero
16.
Exp Parasitol ; 176: 89-98, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28286325

RESUMO

Toxoplasma gondii microneme proteins (TgMICs), secreted by micronemes upon contact with host cells, are reported to play important roles in multiple stages of the T. gondii life cycle, including parasite motility, invasion, intracellular survival, and egress from host cells. Meanwhile, during these processes, TgMICs participate in many protein-protein and protein-carbohydrate interactions, such as undergoing proteolytic maturation, binding to aldolase, engaging the host cell receptors and forming the moving junction (MJ), relying on different types of ectodomains, transmembrane (TM) domains and cytoplasmic domains (CDs). In this review, we summarize the research advances in protein-protein and protein-carbohydrate interactions related to TgMICs, and their intimate associations with corresponding biological processes during T. gondii infection, which will contribute to an improved understanding of the molecular pathogenesis of T. gondii infection, and provide a basis for developing effective control strategies against T. gondii.


Assuntos
Moléculas de Adesão Celular/metabolismo , Proteínas de Protozoários/metabolismo , Toxoplasma/química , Animais , Frutose-Bifosfato Aldolase/metabolismo , Gliceraldeído-3-Fosfato Desidrogenases/metabolismo , Humanos , Domínios e Motivos de Interação entre Proteínas , Proteólise , Transdução de Sinais , Toxoplasma/metabolismo , Toxoplasma/patogenicidade , Virulência
17.
J Biol Chem ; 290(3): 1432-41, 2015 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-25411252

RESUMO

Toxoplasma gondii parasites must actively invade host cells to propagate. Secretory microneme proteins have been shown to be important for both gliding motility and active invasion. MIC2-M2AP is a protein complex that is essential for productive motility and rapid invasion by binding to host cell surface receptors. To investigate the architecture of the MIC2 and M2AP complex, we identified the minimal domains sufficient for interaction and solved the NMR solution structure of the globular domain of M2AP. We found that M2AP adopts a modified galectin fold similar to the C-terminal domain of another microneme protein, MIC1. NMR and immunoprecipitation analyses implicated hydrophobic residues on one face of the M2AP galectin fold in binding to the membrane proximal sixth thrombospondin type I repeat domain of MIC2. Our findings provide a second example of a galectin fold adapted for microneme protein-protein interactions and suggest a conserved strategy for the assembly and folding of diverse protein complexes.


Assuntos
Proteínas de Membrana/química , Proteínas de Protozoários/química , Toxoplasma/química , Animais , Sítios de Ligação , Células CHO , Carboidratos/química , Cricetinae , Cricetulus , Fibroblastos/parasitologia , Galectinas/química , Deleção de Genes , Humanos , Interações Hidrofóbicas e Hidrofílicas , Ligantes , Espectroscopia de Ressonância Magnética , Complexos Multiproteicos/química , Mutação , Ligação Proteica , Conformação Proteica , Dobramento de Proteína , Estrutura Terciária de Proteína , Trombospondinas/química
18.
Exp Parasitol ; 162: 7-17, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26743188

RESUMO

There have been only a few antigen genes of Eimeria brunetti reported up to now. In this study, the gene encoding the microneme protein 2 (EbMIC2) was isolated from oocysts of E. brunetti by RT-PCR and the immunogenicity of recombinant EbMIC2 was observed. The EbMIC2 was cloned into vector pMD19-T for sequencing. The sequence was compared with the published EbMIC2 gene from GenBank revealed homology of the nucleotide sequence and amino acids sequence were 99.43 and 98.63%, respectively. The correct recombinant pMD-EbMIC2 plasmid was inserted into the pET-28a (+) expressing vector and transformed into competent Escherichia coli BL21 cells for expression. The expressed product was analyzed using SDS-PAGE and Western-blot. The results indicated that the recombinant EbMIC2 protein was recognized strongly by serum from naturally infected chicken with E. brunetti. Rat rcEbMIC2 antisera bound to bands of about 36 kDa in the somatic extract of E. brunetti sporozoites. The recombinant plasmid pVAX1-EbMIC2 was constructed and then the efficacies of recombinant plasmid and recombinant protein were evaluated. The results of IgG antibody level and cytokines concentration suggested that recombinant EbMIC2 could increase the IgG antibody level and induce the expressions of cytokines. Animal challenge experiments demonstrated that the recombinant EbMIC2 protein and recombinant plasmid pVAX1-EbMIC2 could significantly increase the average body weight gains, decrease the mean lesion scores and the oocyst outputs of the immunized chickens and presented high anti-coccidial index. All results suggested that EbMIC2 could become an effective candidate for the development of new vaccine against E. brunetti infection.


Assuntos
Galinhas/parasitologia , Coccidiose/veterinária , Surtos de Doenças/veterinária , Eimeria/isolamento & purificação , Doenças das Aves Domésticas/parasitologia , Proteínas de Protozoários/isolamento & purificação , Animais , Antígenos de Protozoários/genética , Antígenos de Protozoários/imunologia , China/epidemiologia , Clonagem Molecular , Coccidiose/epidemiologia , Coccidiose/parasitologia , DNA Complementar/química , DNA Complementar/metabolismo , Eimeria/imunologia , Regulação da Expressão Gênica , Soros Imunes/imunologia , Doenças das Aves Domésticas/epidemiologia , Doenças das Aves Domésticas/prevenção & controle , Proteínas de Protozoários/genética , Proteínas de Protozoários/imunologia , Vacinas Protozoárias/normas , RNA de Protozoário/isolamento & purificação , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Transcrição Reversa , Alinhamento de Sequência/veterinária , Vacinação/veterinária , Vacinas Sintéticas/normas
19.
Traffic ; 14(10): 1053-64, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23834729

RESUMO

The malaria merozoite invades erythrocytes in the vertebrate host. Iterative rounds of asexual intraerythrocytic replication result in disease. Proteases play pivotal roles in erythrocyte invasion, but little is understood about their mode of action. The Plasmodium falciparum malaria merozoite surface sheddase, PfSUB2, is one such poorly characterized example. We have examined the molecular determinants that underlie the mechanisms by which PfSUB2 is trafficked initially to invasion-associated apical organelles (micronemes) and then across the surface of the free merozoite. We show that authentic promoter activity is important for correct localization of PfSUB2, likely requiring canonical features within the intergenic region 5' of the pfsub2 locus. We further demonstrate that trafficking of PfSUB2 beyond an early compartment in the secretory pathway requires autocatalytic protease activity. Finally, we show that the PfSUB2 transmembrane domain is required for microneme targeting, while the cytoplasmic domain is essential for surface translocation of the protease to the parasite posterior following discharge from micronemes. The interplay of pre- and post-translational regulatory elements that coordinate subcellular trafficking of PfSUB2 provides the parasite with exquisite control over enzyme-substrate interactions.


Assuntos
Epitopos/metabolismo , Malária Falciparum/metabolismo , Plasmodium falciparum/metabolismo , Transporte Proteico/fisiologia , Proteínas de Protozoários/metabolismo , Subtilisinas/metabolismo , Epitopos/genética , Epitopos/imunologia , Eritrócitos/imunologia , Eritrócitos/metabolismo , Expressão Gênica/genética , Expressão Gênica/imunologia , Malária Falciparum/genética , Malária Falciparum/imunologia , Merozoítos/imunologia , Merozoítos/metabolismo , Peptídeo Hidrolases/genética , Peptídeo Hidrolases/imunologia , Peptídeo Hidrolases/metabolismo , Plasmodium falciparum/genética , Plasmodium falciparum/imunologia , Regiões Promotoras Genéticas/genética , Regiões Promotoras Genéticas/imunologia , Transporte Proteico/genética , Transporte Proteico/imunologia , Proteólise , Proteínas de Protozoários/genética , Proteínas de Protozoários/imunologia , Subtilisinas/genética , Subtilisinas/imunologia
20.
Sci Rep ; 14(1): 4851, 2024 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-38418588

RESUMO

Eimeria species serve as promising eukaryotic vaccine vectors. And that the location of heterologous antigens in the subcellular components of genetically modified Eimeria may determine the magnitude and type of immune responses. Therefore, our study aimed to target a heterologous fluorescent protein to the cell surface or microneme, two locations where are more effective in inducing protective immunity, of Eimeria tenella and E. acervulina sporozoites. We used an enhanced yellow fluorescent protein (EYFP) as a tagging biomarker, fusing variously with some localization or whole sequences of compartmental proteins for targeting. After acquiring stable transgenic Eimeria populations, we observed EYFP expressing in expected locations with certain strategies. That is, EYFP successfully localized to the surface when it was fused between signal peptides and mature products of surface antigen 1 (SAG1). Furthermore, EYFP was efficiently targeted to the apical end, an optimal location for secretory organelle known as the microneme, when fused to the C terminus of microneme protein 2. Unexpectedly, EYFP exhibited dominantly in the apical end with only weak expression on the surface of the transgenic sporozoites when the parasites were transfected with plasmid with EYFP fused between signal peptides and mature products of E. tenella SAG 13. These strategies worked in both E. tenella and E. acervulina, laying a solid foundation for studying E. tenella and E. acervulina-based live vaccines that can be further tailored to the inclusion of cargo immunogens from other pathogens.


Assuntos
Coccidiose , Eimeria , Parasitos , Doenças das Aves Domésticas , Animais , Coccidiose/parasitologia , Animais Geneticamente Modificados , Sinais Direcionadores de Proteínas , Esporozoítos/metabolismo , Galinhas/parasitologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa