Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
EMBO Mol Med ; 16(4): 723-754, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38514791

RESUMO

Vaccination with infectious Plasmodium falciparum (Pf) sporozoites (SPZ) administered with antimalarial drugs (PfSPZ-CVac), confers superior sterilizing protection against infection when compared to vaccination with replication-deficient, radiation-attenuated PfSPZ. However, the requirement for drug administration constitutes a major limitation for PfSPZ-CVac. To obviate this limitation, we generated late liver stage-arresting replication competent (LARC) parasites by deletion of the Mei2 and LINUP genes (mei2-/linup- or LARC2). We show that Plasmodium yoelii (Py) LARC2 sporozoites did not cause breakthrough blood stage infections and engendered durable sterilizing immunity against various infectious sporozoite challenges in diverse strains of mice. We next genetically engineered a PfLARC2 parasite strain that was devoid of extraneous DNA and produced cryopreserved PfSPZ-LARC2. PfSPZ-LARC2 liver stages replicated robustly in liver-humanized mice but displayed severe defects in late liver stage differentiation and did not form liver stage merozoites. This resulted in complete abrogation of parasite transition to viable blood stage infection. Therefore, PfSPZ-LARC2 is the next-generation vaccine strain expected to unite the safety profile of radiation-attenuated PfSPZ with the superior protective efficacy of PfSPZ-CVac.


Assuntos
Vacinas Antimaláricas , Malária Falciparum , Parasitos , Animais , Camundongos , Plasmodium falciparum/genética , Malária Falciparum/prevenção & controle , Deleção de Genes , Vacinas Antimaláricas/genética , Vacinas Atenuadas/genética , Esporozoítos/genética
2.
Sci Rep ; 13(1): 21071, 2023 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-38030676

RESUMO

The efficacy of pre-erythrocytic stage malaria antigens or vaccine platforms is routinely assessed in murine models challenged with Plasmodium sporozoites. Relative liver-stage parasite burden is quantified using reverse transcription quantitative PCR (RTqPCR), which relies on constitutively expressed endogenous control reference genes. However, the stability of host-reference gene expression for RTqPCR analysis following Plasmodium challenge and immunization has not been systematically evaluated. Herein, we evaluated the stability of expression of twelve common RTqPCR reference genes in a murine model of Plasmodium yoelii sporozoite challenge and DNA-adenovirus IV 'Prime-Target' immunization. Significant changes in expression for six of twelve reference genes were shown by one-way ANOVA, when comparing gene expression levels among challenge, immunized, and naïve mice groups. These changes were attributed to parasite challenge or immunization when comparing group means using post-hoc Bonferroni corrected multiple comparison testing. Succinate dehydrogenase (SDHA) and TATA-binding protein (TBP) were identified as stable host-reference genes suitable for relative RTqPCR data normalisation, using the RefFinder package. We defined a robust threshold of 'partial-protection' with these genes and developed a strategy to simultaneously quantify matched host parasite burden and cytokine responses following immunisation or challenge. This is the first report systematically identifying reliable host reference genes for RTqPCR analysis following Plasmodium sporozoite challenge. A robust RTqPCR protocol incorporating reliable reference genes which enables simultaneous analysis of host whole-liver cytokine responses and parasite burden will significantly standardise and enhance results between international malaria vaccine efficacy studies.


Assuntos
Vacinas Antimaláricas , Malária , Parasitos , Plasmodium yoelii , Animais , Camundongos , Parasitos/genética , Malária/parasitologia , Vacinas Antimaláricas/genética , Imunidade , Citocinas/genética , Expressão Gênica , Esporozoítos/genética , Camundongos Endogâmicos BALB C , Plasmodium yoelii/genética
3.
Malar J ; 20(1): 308, 2021 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-34243763

RESUMO

BACKGROUND: Vaccination with radiation-attenuated Plasmodium falciparum sporozoites is known to induce protective immunity. However, the mechanisms underlying this protection remain unclear. In this work, two recent radiation-attenuated sporozoite vaccination studies were used to identify potential transcriptional correlates of vaccination-induced protection. METHODS: Longitudinal whole blood RNAseq transcriptome responses to immunization with radiation-attenuated P. falciparum sporozoites were analysed and compared across malaria-naïve adult participants (IMRAS) and malaria-experienced adult participants (BSPZV1). Parasite dose and method of delivery differed between trials, and immunization regimens were designed to achieve incomplete protective efficacy. Observed protective efficacy was 55% in IMRAS and 20% in BSPZV1. Study vaccine dosings were chosen to elicit both protected and non-protected subjects, so that protection-associated responses could be identified. RESULTS: Analysis of comparable time points up to 1 week after the first vaccination revealed a shared cross-study transcriptional response programme, despite large differences in number and magnitude of differentially expressed genes between trials. A time-dependent regulatory programme of coherent blood transcriptional modular responses was observed, involving induction of inflammatory responses 1-3 days post-vaccination, with cell cycle responses apparent by day 7 in protected individuals from both trials. Additionally, strongly increased induction of inflammation and interferon-associated responses was seen in non-protected IMRAS participants. All individuals, except for non-protected BSPZV1 participants, showed robust upregulation of cell-cycle associated transcriptional responses post vaccination. CONCLUSIONS: In summary, despite stark differences between the two studies, including route of vaccination and status of malaria exposure, responses were identified that were associated with protection after PfRAS vaccination. These comprised a moderate early interferon response peaking 2 days post vaccination, followed by a later proliferative cell cycle response steadily increasing over the first 7 days post vaccination. Non-protection is associated with deviations from this model, observed in this study with over-induction of early interferon responses in IMRAS and failure to mount a cell cycle response in BSPZV1.


Assuntos
Vacinas Antimaláricas/uso terapêutico , Malária Falciparum/prevenção & controle , Anticorpos Antiprotozoários/sangue , Ensaios Clínicos como Assunto , Humanos , Vacinas Antimaláricas/administração & dosagem , Plasmodium falciparum/genética , Plasmodium falciparum/imunologia , Proteínas de Protozoários/genética , Esporozoítos/genética , Esporozoítos/imunologia , Transcrição Gênica , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/uso terapêutico
4.
Parasitol Res ; 120(7): 2689-2693, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34146126

RESUMO

Poultry coccidiosis causes considerable economical losses to the livestock industry. Eimeria parasites are responsible for this disease. On a global scale, E. acervulina and E. tenella are amongst the most common Eimeria spp. infecting broilers. E. tenella is commonly used as infection model in in vivo and in vitro studies. On the other hand, E. acervulina has barely been studied under in vitro conditions. A well established and widely used in vitro model for E. tenella infection is the Madin-Darby bovine kidney cell line (MDBK); however, little is known regarding suitability of MDBK cells as host cells for E. acervulina. We infected MDBK monolayers with two different doses, 5 × 104 and 2 × 105, of E. acervulina sporozoites and evaluated cultures at 24 and 96 h post infection (hpi). For comparison, we ran an identical infection assay using E. tenella sporozoites. To assess parasite reproduction, the number of DNA copies of E. acervulina SCAR marker and E. tenella ITS-1 gene was quantified using real-time quantitative PCR. We found that the number of E. acervulina copies increased significantly at 24 hpi in comparison to E. tenella (p < 0.05). After 96 hpi, E. acervulina gene copies were considerably reduced while E. tenella continued to multiply (p < 0.05). Our results show that MDBK monolayers could be used for in vitro research aimed to study E. acervulina sporozoite cell invasion. Nevertheless, modifications of in vitro cultivation appear necessary to allow qualitative and quantitative studies over longer periods of parasite reproduction.


Assuntos
Eimeria/fisiologia , Rim/parasitologia , Animais , Bovinos , Linhagem Celular , Galinhas/parasitologia , Coccidiose/parasitologia , Coccidiose/veterinária , Eimeria/classificação , Eimeria/genética , Eimeria tenella/genética , Eimeria tenella/fisiologia , Células Epiteliais , Rim/citologia , Doenças das Aves Domésticas/parasitologia , Reação em Cadeia da Polimerase em Tempo Real , Esporozoítos/classificação , Esporozoítos/genética , Esporozoítos/fisiologia
5.
Exp Parasitol ; 220: 108034, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33188795

RESUMO

Eimeria ninakohlyakimovae represents a highly pathogenic coccidian parasite causing severe haemorrhagic typhlocolitis in goat kids worldwide. NETosis was recently described as an efficient defense mechanism of polymorphonuclear neutrophils (PMN) acting against different parasites in vitro and in vivo. In vitro interactions of caprine PMN with parasitic stages of E. ninakohlyakimovae (i. e. oocysts and sporozoites) as well as soluble oocyst antigens (SOA) were analyzed at different ratios, concentrations and time spans. Extracellular DNA staining was used to illustrate classical molecules induced during caprine NETosis [i. e. histones (H3) and neutrophil elastase (NE)] via antibody-based immunofluorescence analyses. Functional inhibitor treatments with DPI and DNase I were applied to unveil role of NADPH oxidase (NOX) and characterize DNA-backbone composition of E. ninakohlyakimovae-triggered caprine NETosis. Scanning electron microscopy (SEM)- and immunofluorescence-analyses demonstrated that caprine PMN underwent NETosis upon contact with sporozoites and oocysts of E. ninakohlyakimovae, ensnaring filaments which firmly entrapped parasites. Detailed co-localization studies of E. ninakohlyakimovae-induced caprine NETosis revealed presence of PMN-derived DNA being adorned with nuclear H3 and NE corroborating molecular characteristics of NETosis. E. ninakohlyakoimovae-induced caprine NETosis was found to be NOX-independent since DPI inhibition led to a slight decrease of NETosis. Exposure of caprine PMN to vital E. ninakohlyakimovae sporozoites as well as SOA resulted in up-regulation of IL-12, TNF-α, IL-6, CCL2 and iNOS gene transcription in stimulated PMN. Since vital E. ninakohlyakimovae-sporozoites induced caprine NETosis, this effective entrapment mechanism might reduce initial sporozoite epithelial host cell invasion during goat coccidiosis ultimately resulting in less macromeront formation and reduced merozoites I production.


Assuntos
Coccidiose/veterinária , Citocinas/genética , Eimeria/imunologia , Doenças das Cabras/parasitologia , Neutrófilos/parasitologia , Análise de Variância , Animais , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Coccidiose/imunologia , Coccidiose/parasitologia , Colite/parasitologia , Colite/veterinária , Citocinas/metabolismo , Eimeria/genética , Eimeria/ultraestrutura , Hemorragia Gastrointestinal/parasitologia , Hemorragia Gastrointestinal/veterinária , Doenças das Cabras/imunologia , Cabras , Interleucina-12/genética , Interleucina-12/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Microscopia Eletrônica de Varredura/veterinária , NADPH Oxidases/metabolismo , Neutrófilos/imunologia , Neutrófilos/ultraestrutura , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Oocistos/genética , Oocistos/imunologia , Reação em Cadeia da Polimerase/veterinária , Esporozoítos/genética , Esporozoítos/imunologia , Transcrição Gênica , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , Tiflite/parasitologia , Tiflite/veterinária , Regulação para Cima
6.
mBio ; 11(1)2020 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-32019802

RESUMO

The apicomplexan parasites Plasmodium spp. are the causative agents of malaria, a disease that poses a significant global health burden. Plasmodium spp. initiate infection of the human host by transforming and replicating within hepatocytes. This liver stage (LS) is poorly understood compared to other Plasmodium life stages, which has hindered our ability to target these parasites for disease prevention. We conducted an extensive transcriptome sequencing (RNA-Seq) analysis throughout the Plasmodium berghei LS, covering as early as 2 h postinfection (hpi) and extending to 48 hpi. Our data revealed that hundreds of genes are differentially expressed at 2 hpi and that multiple genes shown to be important for later infection are upregulated as early as 12 hpi. Using hierarchical clustering along with coexpression analysis, we identified clusters functionally enriched for important liver-stage processes such as interactions with the host cell and redox homeostasis. Furthermore, some of these clusters were highly correlated to the expression of ApiAP2 transcription factors, while showing enrichment of mostly uncharacterized DNA binding motifs. This finding indicates potential LS targets for these transcription factors, while also hinting at alternative uncharacterized DNA binding motifs and transcription factors during this stage. Our work presents a window into the previously undescribed transcriptome of Plasmodium upon host hepatocyte infection to enable a comprehensive view of the parasite's LS. These findings also provide a blueprint for future studies that extend hypotheses concerning LS gene function in P. berghei to human-infective Plasmodium parasites.IMPORTANCE The LS of Plasmodium infection is an asymptomatic yet necessary stage for producing blood-infective parasites, the causative agents of malaria. Blocking the liver stage of the life cycle can prevent clinical malaria, but relatively less is known about the parasite's biology at this stage. Using the rodent model P. berghei, we investigated whole-transcriptome changes occurring as early as 2 hpi of hepatocytes. The transcriptional profiles of early time points (2, 4, 12, and 18 hpi) have not been accessible before due to the technical challenges associated with liver-stage infections. Our data now provide insights into these early parasite fluxes that may facilitate establishment of infection, transformation, and replication in the liver.


Assuntos
Perfilação da Expressão Gênica , Hepatócitos/parasitologia , Fígado/parasitologia , Malária/parasitologia , Plasmodium berghei/genética , Células Hep G2 , Interações Hospedeiro-Parasita/genética , Humanos , Estágios do Ciclo de Vida , Plasmodium berghei/fisiologia , Proteínas de Protozoários/genética , RNA-Seq , Esporozoítos/genética , Esporozoítos/fisiologia
7.
Nucleic Acids Res ; 48(5): 2303-2311, 2020 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-32034421

RESUMO

Chromatin conformation assays such as Hi-C cannot directly measure differences in 3D architecture between cell types or cell states. For this purpose, two or more Hi-C experiments must be carried out, but direct comparison of the resulting Hi-C matrices is confounded by several features of Hi-C data. Most notably, the genomic distance effect, whereby contacts between pairs of genomic loci that are proximal along the chromosome exhibit many more Hi-C contacts that distal pairs of loci, dominates every Hi-C matrix. Furthermore, the form that this distance effect takes often varies between different Hi-C experiments, even between replicate experiments. Thus, a statistical confidence measure designed to identify differential Hi-C contacts must accurately account for the genomic distance effect or risk being misled by large-scale but artifactual differences. ACCOST (Altered Chromatin COnformation STatistics) accomplishes this goal by extending the statistical model employed by DEseq, re-purposing the 'size factors,' which were originally developed to account for differences in read depth between samples, to instead model the genomic distance effect. We show via analysis of simulated and real data that ACCOST provides unbiased statistical confidence estimates that compare favorably with competing methods such as diffHiC, FIND and HiCcompare. ACCOST is freely available with an Apache license at https://bitbucket.org/noblelab/accost.


Assuntos
Cromatina/química , DNA/química , Loci Gênicos , Genoma , Software , Animais , Linhagem Celular , Cromatina/metabolismo , DNA/metabolismo , Epistasia Genética , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Humanos , Linfócitos/citologia , Linfócitos/metabolismo , Camundongos , Conformação Molecular , Plasmodium falciparum/genética , Esporozoítos/genética , Trofozoítos/genética
8.
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
9.
Sci Rep ; 9(1): 8386, 2019 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-31182757

RESUMO

A highly effective vaccine that confers sterile protection to malaria is urgently needed. Immunization under chemoprophylaxis with sporozoites (CPS) consistently confers high levels of protection in the Controlled Human Malaria infection (CHMI) model. To provide a broad, unbiased assessment of the composition and kinetics of direct ex vivo human immune responses to CPS, we profiled whole-blood transcriptomes by RNA-seq before and during CPS immunization and following CHMI challenge. Differential expression of genes enriched in modules related to T cells, NK cells, protein synthesis, and mitochondrial processes were detected in fully protected individuals four weeks after the first immunization. Non-protected individuals demonstrated transcriptomic changes after the third immunization and the day of treatment, with upregulation of interferon and innate inflammatory genes and downregulation of B-cell signatures. Protected individuals demonstrated more significant interactions between blood transcription modules compared to non-protected individuals several weeks after the second and third immunizations. These data provide insight into the molecular and cellular basis of CPS-induced immune protection from P. falciparum infection.


Assuntos
Cloroquina/farmacologia , Malária Falciparum/tratamento farmacológico , Plasmodium falciparum/genética , Transcriptoma/efeitos dos fármacos , Animais , Antimaláricos/farmacologia , Linfócitos B/efeitos dos fármacos , Linfócitos B/metabolismo , Quimioprevenção , Humanos , Vacinas Antimaláricas/genética , Vacinas Antimaláricas/farmacologia , Malária Falciparum/genética , Malária Falciparum/metabolismo , Malária Falciparum/parasitologia , Plasmodium falciparum/patogenicidade , Esporozoítos/efeitos dos fármacos , Esporozoítos/genética , Esporozoítos/patogenicidade , Transcriptoma/genética , Vacinação
10.
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
11.
PLoS One ; 13(10): e0204047, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30303978

RESUMO

Theileria parva is a protozoan parasite transmitted by the brown ear tick Rhipicephalus appendiculatus that causes East Coast fever (ECF) in cattle, resulting in substantial economic losses in the regions of southern, eastern and central Africa. The schizont form of the parasite transforms the bovine host lymphocytes into actively proliferating cancer-like cells. However, how T. parva causes bovine host cells to proliferate and maintain a cancerous phenotype following infection is still poorly understood. On the other hand, current efforts to develop improved vaccines have identified only a few candidate antigens. In the present paper, we report the first comparative transcriptomic analysis throughout the course of T. parva infection. We observed that the development of sporoblast into sporozoite and then the establishment in the host cells as schizont is accompanied by a drastic increase of upregulated genes in the schizont stage of the parasite. In contrast, the ten highest gene expression values occurred in the arthropod vector stages. A comparative analysis showed that 2845 genes were upregulated in both sporozoite and schizont stages compared to the sporoblast. In addition, 647 were upregulated only in the sporozoite whereas 310 were only upregulated in the schizont. We detected low p67 expression in the schizont stage, an unexpected finding considering that p67 has been reported as a sporozoite stage-specific gene. In contrast, we found that transcription of p67 was 20 times higher in the sporoblast than in the sporozoite. Using the expression profiles of recently identified candidate vaccine antigens as a benchmark for selection for novel potential vaccine candidates, we identified three genes with expression similar to p67 and several other genes similar to Tp1-Tp10 schizont vaccine antigens. We propose that the antigenicity or chemotherapeutic potential of this panel of new candidate antigens be further investigated. Structural comparisons of the transcripts generated here with the existing gene models for the respective loci revealed indels. Our findings can be used to improve the structural annotation of the T. parva genome, and the identification of alternatively spliced transcripts.


Assuntos
Antígenos de Protozoários/genética , Perfilação da Expressão Gênica/métodos , Theileria parva/crescimento & desenvolvimento , Theileriose/parasitologia , Animais , Antígenos de Protozoários/imunologia , Bovinos , Regulação da Expressão Gênica no Desenvolvimento , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Proteínas de Protozoários/genética , Proteínas de Protozoários/imunologia , Vacinas Protozoárias/genética , Vacinas Protozoárias/imunologia , Esquizontes/genética , Esquizontes/imunologia , Análise de Sequência de RNA/métodos , Esporozoítos/genética , Esporozoítos/imunologia , Theileria parva/genética , Theileria parva/imunologia , Regulação para Cima
12.
Am J Trop Med Hyg ; 99(4): 827-832, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30141395

RESUMO

In August 2017, the National Institute of Allergy and Infectious Diseases convened a meeting, entitled "Understanding the Liver-Stage Biology of Malaria Parasites to Enable and Accelerate the Development of a Highly Efficacious Vaccine," to discuss the needs and strategies to develop a highly efficacious, whole organism-based vaccine targeting the liver stage of malaria parasites. It was concluded that attenuated sporozoite platforms have proven to be promising approaches, and that late-arresting sporozoites could potentially offer greater vaccine performance than early-arresting sporozoites against malaria. New knowledge and emerging technologies have made the development of late-arresting sporozoites feasible. Highly integrated approaches involving liver-stage research, "omics" studies, and cutting-edge genetic editing technologies, combined with in vitro culture systems or unique animal models, are needed to accelerate the discovery of candidates for a late-arresting, genetically attenuated parasite vaccine.


Assuntos
Fígado/imunologia , Vacinas Antimaláricas/genética , Malária Falciparum/prevenção & controle , Malária Vivax/prevenção & controle , Plasmodium falciparum/imunologia , Plasmodium vivax/imunologia , Esporozoítos/imunologia , Animais , Modelos Animais de Doenças , Raios gama , Engenharia Genética/métodos , Humanos , Fígado/parasitologia , Malária/imunologia , Malária/parasitologia , Malária/prevenção & controle , Vacinas Antimaláricas/administração & dosagem , Vacinas Antimaláricas/metabolismo , Malária Falciparum/imunologia , Malária Falciparum/parasitologia , Malária Vivax/imunologia , Malária Vivax/parasitologia , Camundongos , Plasmodium berghei/química , Plasmodium berghei/genética , Plasmodium berghei/imunologia , Plasmodium berghei/efeitos da radiação , Plasmodium falciparum/química , Plasmodium falciparum/genética , Plasmodium falciparum/efeitos da radiação , Plasmodium vivax/química , Plasmodium vivax/genética , Plasmodium vivax/efeitos da radiação , Plasmodium yoelii/química , Plasmodium yoelii/genética , Plasmodium yoelii/imunologia , Plasmodium yoelii/efeitos da radiação , Esporozoítos/química , Esporozoítos/genética , Esporozoítos/efeitos da radiação , Vacinas Atenuadas
13.
Parasitol Res ; 117(8): 2487-2497, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29797085

RESUMO

Malaria is transmitted through the injection of Plasmodium sporozoites into the skin by Anopheles mosquitoes. The parasites first replicate within the liver before infecting red blood cells, which leads to the symptoms of the disease. Experimental immunization with attenuated sporozoites that arrest their development in the liver has been extensively investigated in rodent models and humans. Recent technological advances have included the capacity to cryopreserve sporozoites for injection, which has enabled a series of controlled studies on human infection with sporozoites. Here, we used a cryopreservation protocol to test the efficiency of genetically attenuated cryopreserved sporozoites for immunization of mice in comparison with freshly isolated controls. This showed that cryopreserved sporozoites are highly viable as judged by their capacity to migrate in vitro but show only 20% efficiency in liver infection, which impacts their capacity to generate protection of animals in immunization experiments.


Assuntos
Malária/prevenção & controle , Plasmodium berghei/imunologia , Esporozoítos/imunologia , Vacinação , Vacinas Atenuadas/imunologia , Animais , Anopheles/parasitologia , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Criopreservação , Células Hep G2 , Humanos , Fígado/parasitologia , Malária/parasitologia , Camundongos , Camundongos Endogâmicos C57BL , Plasmodium berghei/genética , Esporozoítos/genética , Esporozoítos/metabolismo
14.
JCI Insight ; 2(1): e89154, 2017 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-28097230

RESUMO

BACKGROUND: A radiation-attenuated Plasmodium falciparum (Pf) sporozoite (SPZ) malaria vaccine, PfSPZ Vaccine, protected 6 of 6 subjects (100%) against homologous Pf (same strain as in the vaccine) controlled human malaria infection (CHMI) 3 weeks after 5 doses administered intravenously. The next step was to assess protective efficacy against heterologous Pf (different from Pf in the vaccine), after fewer doses, and at 24 weeks. METHODS: The trial assessed tolerability, safety, immunogenicity, and protective efficacy of direct venous inoculation (DVI) of 3 or 5 doses of PfSPZ Vaccine in non-immune subjects. RESULTS: Three weeks after final immunization, 5 doses of 2.7 × 105 PfSPZ protected 12 of 13 recipients (92.3% [95% CI: 48.0, 99.8]) against homologous CHMI and 4 of 5 (80.0% [10.4, 99.5]) against heterologous CHMI; 3 doses of 4.5 × 105 PfSPZ protected 13 of 15 (86.7% [35.9, 98.3]) against homologous CHMI. Twenty-four weeks after final immunization, the 5-dose regimen protected 7 of 10 (70.0% [17.3, 93.3]) against homologous and 1 of 10 (10.0% [-35.8, 45.6]) against heterologous CHMI; the 3-dose regimen protected 8 of 14 (57.1% [21.5, 76.6]) against homologous CHMI. All 22 controls developed Pf parasitemia. PfSPZ Vaccine was well tolerated, safe, and easy to administer. No antibody or T cell responses correlated with protection. CONCLUSIONS: We have demonstrated for the first time to our knowledge that PfSPZ Vaccine can protect against a 3-week heterologous CHMI in a limited group of malaria-naive adult subjects. A 3-dose regimen protected against both 3-week and 24-week homologous CHMI (87% and 57%, respectively) in this population. These results provide a foundation for developing an optimized immunization regimen for preventing malaria. TRIAL REGISTRATION: ClinicalTrials.gov NCT02215707. FUNDING: Support was provided through the US Army Medical Research and Development Command, Military Infectious Diseases Research Program, and the Naval Medical Research Center's Advanced Medical Development Program.


Assuntos
Malária Falciparum/terapia , Plasmodium falciparum/efeitos dos fármacos , Esporozoítos/efeitos dos fármacos , Vacinas Atenuadas/administração & dosagem , Administração Intravenosa , Adulto , Feminino , Humanos , Malária Falciparum/prevenção & controle , Masculino , Plasmodium falciparum/genética , Esporozoítos/genética , Linfócitos T/imunologia , Vacinas Atenuadas/uso terapêutico , Sequenciamento Completo do Genoma/métodos
15.
Sci Rep ; 6: 38170, 2016 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-27901110

RESUMO

It has been reported that non-selective autophagy of infected hepatocytes could facilitate the development of malaria in the liver stage, but the fate of parasites following selective autophagy of infected hepatocytes is still not very clear. Here, we confirmed that sporozoite infection can induce a selective autophagy-like process targeting EEFs (exo-erythrocytic forms) in Hepa1-6. Rapamycin treatment greatly enhanced this process in EEFs and non-selective autophagy of infected Hepa1-6 cells and enhanced the development of the malaria liver stage in vivo. Although rapamycin promoted the fusion of autophagosomes containing the malaria parasite with lysosomes, some parasites inside the autophagosome survived and replicated normally. Further study showed that the maturation of affected autolysosomes was greatly inhibited. Therefore, in addition to the previously described positive role of rapamycin-induced nonselective autophagy of hepatocytes, we provide evidence that the survival of EEFs in the autophagosome of the infected hepatocytes also contributes to rapamycin-enhanced development of the malaria liver stage, possibly due to the suppression of autolysosome maturation by EEFs. These data suggest that the inhibition of autolysosome maturation might be a novel escape strategy used by the malaria liver stage.


Assuntos
Autofagossomos/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Malária/metabolismo , Sirolimo/farmacologia , Animais , Antibióticos Antineoplásicos/farmacologia , Autofagossomos/metabolismo , Autofagossomos/parasitologia , Autofagia/efeitos dos fármacos , Carcinoma Hepatocelular/parasitologia , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Hepatócitos/metabolismo , Hepatócitos/parasitologia , Interações Hospedeiro-Parasita/efeitos dos fármacos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/parasitologia , Neoplasias Hepáticas/parasitologia , Neoplasias Hepáticas/patologia , Malária/parasitologia , Camundongos , Plasmodium yoelii/efeitos dos fármacos , Plasmodium yoelii/crescimento & desenvolvimento , Plasmodium yoelii/fisiologia , Esporozoítos/genética , Esporozoítos/fisiologia
16.
J Exp Med ; 202(2): 225-30, 2005 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-16027235

RESUMO

The Plasmodium life cycle is a sequence of alternating invasive and replicative stages within the vertebrate and invertebrate hosts. How malarial parasites exit their host cells after completion of reproduction remains largely unsolved. Inhibitor studies indicated a role of Plasmodium cysteine proteases in merozoite release from host erythrocytes. To validate a vital function of malarial cysteine proteases in active parasite egress, we searched for target genes that can be analyzed functionally by reverse genetics. Herein, we describe a complete arrest of Plasmodium sporozoite egress from Anopheles midgut oocysts by targeted disruption of a stage-specific cysteine protease. Our findings show that sporozoites exit oocysts by parasite-dependent proteolysis rather than by passive oocyst rupture resulting from parasite growth. We provide genetic proof that malarial cysteine proteases are necessary for egress of invasive stages from their intracellular compartment and propose that similar cysteine protease-dependent mechanisms occur during egress from liver-stage and blood-stage schizonts.


Assuntos
Cisteína Endopeptidases/metabolismo , Oocistos/fisiologia , Plasmodium berghei/fisiologia , Proteínas de Protozoários/metabolismo , Esporozoítos/fisiologia , Animais , Animais Geneticamente Modificados , Anopheles/parasitologia , Cisteína Endopeptidases/genética , Oocistos/genética , Plasmodium berghei/genética , Proteínas de Protozoários/genética , Esporozoítos/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA