Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
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
2.
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
3.
Parasitol Int ; 85: 102435, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34390881

RESUMO

Malaria remains a heavy global burden on human health, and it is important to understand the molecular and cellular biology of the parasite to find targets for drug and vaccine development. The mouse malaria model is an essential tool to characterize the function of identified molecules; however, robust technologies for targeted gene deletions are still poorly developed for the widely used rodent malaria parasite, Plasmodium yoelii. To overcome this problem, we established a DiCre-loxP inducible knockout (iKO) system in P. yoelii, which showed more than 80% excision efficacy of the target locus and more than 90% reduction of locus transcripts 24 h (one cell cycle) after RAP administration. Using this developed system, cAMP-dependent protein kinase (PKAc) was inducibly disrupted and the phenotypes of the resulting PKAc-iKO parasites were analyzed. We found that PKAc-iKO parasites showed severe growth and erythrocyte invasion defects. We also found that disruption of PKAc impaired the secretion of AMA1 in P. yoelii, in contrast to a report showing no role of PKAc in AMA1 secretion in P. falciparum. This discrepancy may be related to the difference in the timing of AMA1 distribution to the merozoite surface, which occurs just after egress for P. falciparum, but after several minutes for P. yoelii. Secretions of PyEBL, Py235, and RON2 were not affected by the disruption of PKAc in P. yoelii. PyRON2 was already secreted to the merozoite surface immediately after merozoite egress, which is inconsistent with the current model that RON2 is injected into the erythrocyte cytosol. Further investigations are required to understand the role of RON2 exposed on the merozoite surface.


Assuntos
Antígenos de Protozoários/biossíntese , Proteínas Quinases Dependentes de AMP Cíclico/genética , Proteínas de Membrana/biossíntese , Plasmodium yoelii/genética , Proteínas de Protozoários/genética , Animais , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Feminino , Merozoítos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos ICR , Plasmodium yoelii/enzimologia , Plasmodium yoelii/metabolismo , Proteínas de Protozoários/biossíntese , Proteínas de Protozoários/metabolismo
4.
J Exp Med ; 217(2)2020 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-31748243

RESUMO

CD4+ T follicular helper (Tfh) cells dominate the acute response to a blood-stage Plasmodium infection and provide signals to direct B cell differentiation and protective antibody expression. We studied antigen-specific CD4+ Tfh cells responding to Plasmodium infection in order to understand the generation and maintenance of the Tfh response. We discovered that a dominant, phenotypically stable, CXCR5+ Tfh population emerges within the first 4 d of infection and results in a CXCR5+ CCR7+ Tfh/central memory T cell response that persists well after parasite clearance. We also found that CD4+ T cell priming by B cells was both necessary and sufficient to generate this Tfh-dominant response, whereas priming by conventional dendritic cells was dispensable. This study provides important insights into the development of CD4+ Tfh cells during Plasmodium infection and highlights the heterogeneity of antigen-presenting cells involved in CD4+ T cell priming.


Assuntos
Linfócitos B/imunologia , Malária/imunologia , Plasmodium yoelii/genética , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/imunologia , Células Cultivadas , Células Dendríticas/imunologia , Epitopos de Linfócito T/genética , Epitopos de Linfócito T/imunologia , Memória Imunológica , Ativação Linfocitária/imunologia , Malária/parasitologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Organismos Geneticamente Modificados , Fenótipo , Receptores CCR7/metabolismo , Receptores CXCR5/metabolismo
5.
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
6.
Vaccine ; 35(24): 3171-3177, 2017 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-28483194

RESUMO

A CD1d-binding, invariant (i) natural killer T (NKT)-cell stimulatory glycolipid, α-Galactosylceramide (αGalCer), has been shown to act as an adjuvant. We previously identified a fluorinated phenyl ring-modified αGalCer analog, 7DW8-5, displaying a higher binding affinity for CD1d molecule and more potent adjuvant activity than αGalCer. In the present study, 7DW8-5 co-administered intramuscularly (i.m.) with a recombinant adenovirus expressing a Plasmodium yoelii circumsporozoite protein (PyCSP), AdPyCS, has led to a co-localization of 7DW8-5 and a PyCSP in draining lymph nodes (dLNs), particularly in dendritic cells (DCs). This occurrence initiates a cascade of events, such as the recruitment of DCs to dLNs and their activation and maturation, and the enhancement of the ability of DCs to prime CD8+ T cells induced by AdPyCS and ultimately leading to a potent adjuvant effect and protection against malaria.


Assuntos
Adenoviridae/genética , Adjuvantes Imunológicos , Antígenos CD1d/imunologia , Galactosilceramidas/imunologia , Vacinas Antimaláricas/imunologia , Animais , Antígenos CD1d/metabolismo , Antígenos de Protozoários/administração & dosagem , Antígenos de Protozoários/genética , Antígenos de Protozoários/imunologia , Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Galactosilceramidas/química , Galactosilceramidas/metabolismo , Imunogenicidade da Vacina , Injeções Intramusculares , Interferon gama/imunologia , Células Matadoras Naturais/imunologia , Ativação Linfocitária , Malária/imunologia , Malária/prevenção & controle , Vacinas Antimaláricas/administração & dosagem , Camundongos , Células T Matadoras Naturais/imunologia , Plasmodium yoelii/química , Plasmodium yoelii/genética , Plasmodium yoelii/imunologia , Vacinas Sintéticas/imunologia
7.
PLoS One ; 11(4): e0154819, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27128437

RESUMO

A malaria vaccine is a public health priority. In order to produce an effective vaccine, a multistage approach targeting both the blood and the liver stage infection is desirable. The vaccine candidates also need to induce balanced immune responses including antibodies, CD4+ and CD8+ T cells. Protein-based subunit vaccines like RTS,S are able to induce strong antibody response but poor cellular reactivity. Adenoviral vectors have been effective inducing protective CD8+ T cell responses in several models including malaria; nonetheless this vaccine platform exhibits a limited induction of humoral immune responses. Two approaches have been used to improve the humoral immunogenicity of recombinant adenovirus vectors, the use of heterologous prime-boost regimens with recombinant proteins or the genetic modification of the hypervariable regions (HVR) of the capsid protein hexon to express B cell epitopes of interest. In this study, we describe the development of capsid modified Ad5 vectors that express a promiscuous Plasmodium yoelii T helper epitope denominated PyT53 within the hexon HVR2 region. Several regimens were tested in mice to determine the relevance of the hexon modification in enhancing protective immune responses induced by the previously described protein-based multi-stage experimental vaccine PyCMP. A heterologous prime-boost immunization regime that combines a hexon modified vector with transgenic expression of PyCMP followed by protein immunizations resulted in the induction of robust antibody and cellular immune responses in comparison to a similar regimen that includes a vector with unmodified hexon. These differences in immunogenicity translated into a better protective efficacy against both the hepatic and red blood cell stages of P. yoelii. To our knowledge, this is the first time that a hexon modification is used to deliver a promiscuous T cell epitope. Our data support the use of such modification to enhance the immunogenicity and protective efficacy of adenoviral based malaria vaccines.


Assuntos
Proteínas do Capsídeo/imunologia , Epitopos de Linfócito T/imunologia , Vacinas Antimaláricas/imunologia , Plasmodium yoelii/imunologia , Adenovírus Humanos/genética , Adenovírus Humanos/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Antiprotozoários/biossíntese , Proteínas do Capsídeo/genética , Citocinas/biossíntese , Epitopos de Linfócito T/genética , Feminino , Vetores Genéticos , Humanos , Imunidade Celular , Malária/imunologia , Malária/parasitologia , Malária/prevenção & controle , Vacinas Antimaláricas/administração & dosagem , Vacinas Antimaláricas/genética , Camundongos , Plasmodium yoelii/genética , Receptor de Morte Celular Programada 1/metabolismo , Proteínas de Protozoários/administração & dosagem , Proteínas de Protozoários/genética , Proteínas de Protozoários/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia
8.
Infect Immun ; 83(10): 3781-92, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26169268

RESUMO

Malaria remains a world-threatening disease largely because of the lack of a long-lasting and fully effective vaccine. MAEBL is a type 1 transmembrane molecule with a chimeric cysteine-rich ectodomain homologous to regions of the Duffy binding-like erythrocyte binding protein and apical membrane antigen 1 (AMA1) antigens. Although MAEBL does not appear to be essential for the survival of blood-stage forms, ectodomains M1 and M2, homologous to AMA1, seem to be involved in parasite attachment to erythrocytes, especially M2. MAEBL is necessary for sporozoite infection of mosquito salivary glands and is expressed in liver stages. Here, the Plasmodium yoelii MAEBL-M2 domain was expressed in a prokaryotic vector. C57BL/6J mice were immunized with doses of P. yoelii recombinant protein rPyM2-MAEBL. High levels of antibodies, with balanced IgG1 and IgG2c subclasses, were achieved. rPyM2-MAEBL antisera were capable of recognizing the native antigen. Anti-MAEBL antibodies recognized different MAEBL fragments expressed in CHO cells, showing stronger IgM and IgG responses to the M2 domain and repeat region, respectively. After a challenge with P. yoelii YM (lethal strain)-infected erythrocytes (IE), up to 90% of the immunized animals survived and a reduction of parasitemia was observed. Moreover, splenocytes harvested from immunized animals proliferated in a dose-dependent manner in the presence of rPyM2-MAEBL. Protection was highly dependent on CD4(+), but not CD8(+), T cells toward Th1. rPyM2-MAEBL antisera were also able to significantly inhibit parasite development, as observed in ex vivo P. yoelii erythrocyte invasion assays. Collectively, these findings support the use of MAEBL as a vaccine candidate and open perspectives to understand the mechanisms involved in protection.


Assuntos
Vacinas Antimaláricas/imunologia , Malária/prevenção & controle , Plasmodium yoelii/imunologia , Proteínas de Protozoários/química , Proteínas de Protozoários/imunologia , Animais , Anticorpos Antiprotozoários/imunologia , Eritrócitos/parasitologia , Feminino , Humanos , Imunização , Malária/imunologia , Malária/mortalidade , Malária/parasitologia , Vacinas Antimaláricas/administração & dosagem , Vacinas Antimaláricas/química , Vacinas Antimaláricas/genética , Masculino , Merozoítos/química , Merozoítos/crescimento & desenvolvimento , Merozoítos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Plasmodium yoelii/química , Plasmodium yoelii/genética , Plasmodium yoelii/crescimento & desenvolvimento , Estrutura Terciária de Proteína , Proteínas de Protozoários/administração & dosagem , Proteínas de Protozoários/genética , Esporozoítos/química , Esporozoítos/crescimento & desenvolvimento , Esporozoítos/imunologia
9.
Cell Microbiol ; 16(10): 1533-48, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24798694

RESUMO

Plasmodium sporozoites are transmitted by Anopheles mosquitoes and first infect the liver of their mammalian host, where they develop as liver stages before the onset of erythrocytic infection and malaria symptoms. Sporozoite entry into hepatocytes is an attractive target for anti-malarial prophylactic strategies but remains poorly understood at the molecular level. Apicomplexan parasites invade host cells by forming a parasitophorous vacuole that is essential for parasite development, a process that involves secretion of apical organelles called rhoptries. We previously reported that the host membrane protein CD81 is required for infection by Plasmodium falciparum and Plasmodium yoelii sporozoites. CD81 acts at an early stage of infection, possibly at the entry step, but the mechanisms involved are still unknown. To investigate the role of CD81 during sporozoite entry, we generated transgenic P. yoelii parasites expressing fluorescent versions of three known rhoptry proteins, RON2, RON4 and RAP2/3. We observed that RON2 and RON4 are lost following rhoptry discharge during merozoite and sporozoite entry. In contrast, our data indicate that RAP2/3 is secreted into the parasitophorous vacuole during infection. We further show that sporozoite rhoptry discharge occurs only in the presence of CD81, providing the first direct evidence for a role of CD81 during sporozoite productive invasion.


Assuntos
Interações Hospedeiro-Parasita/fisiologia , Plasmodium yoelii/patogenicidade , Proteínas de Protozoários/metabolismo , Esporozoítos/patologia , Tetraspanina 28/metabolismo , Animais , Linhagem Celular , Feminino , Proteínas de Fluorescência Verde/genética , Células Hep G2 , Hepatócitos/parasitologia , Humanos , Proteínas Luminescentes/genética , Malária , Camundongos , Camundongos Endogâmicos BALB C , Organismos Geneticamente Modificados , Plasmodium yoelii/genética , Proteínas de Protozoários/biossíntese , Proteínas de Protozoários/genética , Vacúolos/patologia , Proteína Vermelha Fluorescente
10.
Gene Ther ; 21(2): 212-8, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24352195

RESUMO

Dendritic cells (DC) targeting vaccines require high efficiency for uptake, followed by DC activation and maturation. We used magnetic vectors comprising polyethylenimine (PEI)-coated superparamagnetic iron oxide nanoparticles, with hyaluronic acid (HA) of different molecular weights (<10 and 900 kDa) to reduce cytotoxicity and to facilitate endocytosis of particles into DCs via specific surface receptors. DNA encoding Plasmodium yoelii merozoite surface protein 1-19 and a plasmid encoding yellow fluorescent gene were added to the magnetic complexes with various % charge ratios of HA: PEI. The presence of magnetic fields significantly enhanced DC transfection and maturation. Vectors containing a high-molecular-weight HA with 100% charge ratio of HA: PEI yielded a better transfection efficiency than others. This phenomenon was attributed to their longer molecular chains and higher mucoadhesive properties aiding DNA condensation and stability. Insights gained should improve the design of more effective DNA vaccine delivery systems.


Assuntos
Compostos Férricos/metabolismo , Ácido Hialurônico/farmacologia , Polietilenoimina/química , Polietilenoimina/metabolismo , Vacinas de DNA/administração & dosagem , Vacinas de DNA/química , Animais , Antígenos de Protozoários/genética , Células Dendríticas , Compostos Férricos/química , Vetores Genéticos , Fenômenos Magnéticos , Camundongos Endogâmicos C57BL , Nanopartículas/administração & dosagem , Nanopartículas/efeitos adversos , Nanopartículas/química , Plasmodium yoelii/genética , Transfecção
11.
PLoS One ; 7(9): e44943, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22984589

RESUMO

Viral vectored vaccines have been shown to induce both T cell and antibody responses in animals and humans. However, the induction of even higher level T cell responses may be crucial in achieving vaccine efficacy against difficult disease targets, especially in humans. Here we investigate the oligomerization domain of the α-chain of C4b-binding protein (C4 bp) as a candidate T cell "molecular adjuvant" when fused to malaria antigens expressed by human adenovirus serotype 5 (AdHu5) vectored vaccines in BALB/c mice. We demonstrate that i) C-terminal fusion of an oligomerization domain can enhance the quantity of antigen-specific CD4(+) and CD8(+) T cell responses induced in mice after only a single immunization of recombinant AdHu5, and that the T cells maintain similar functional cytokine profiles; ii) an adjuvant effect is observed for AdHu5 vectors expressing either the 42 kDa C-terminal domain of Plasmodium yoelii merozoite surface protein 1 (PyMSP1(42)) or the 83 kDa ectodomain of P. falciparum strain 3D7 apical membrane antigen 1 (PfAMA1), but not a candidate 128kDa P. falciparum MSP1 biallelic fusion antigen; iii) following two homologous immunizations of AdHu5 vaccines, antigen-specific T cell responses are further enhanced, however, in both BALB/c mice and New Zealand White rabbits no enhancement of functional antibody responses is observed; and iv) that the T cell adjuvant activity of C4 bp is not dependent on a functional Fc-receptor γ-chain in the host, but is associated with the oligomerization of small (<80 kDa) antigens expressed by recombinant AdHu5. The oligomerization domain of C4 bp can thus adjuvant T cell responses induced by AdHu5 vectors against selected antigens and its clinical utility as well as mechanism of action warrant further investigation.


Assuntos
Adenoviridae/genética , Proteína de Ligação ao Complemento C4b/metabolismo , Malária/prevenção & controle , Linfócitos T/citologia , Adjuvantes Imunológicos/genética , Animais , Antígenos de Protozoários/genética , Feminino , Vetores Genéticos , Vacinas Antimaláricas/genética , Proteína 1 de Superfície de Merozoito/genética , Camundongos , Camundongos Endogâmicos BALB C , Plasmodium falciparum/genética , Plasmodium yoelii/genética , Estrutura Terciária de Proteína , Coelhos , Receptores de IgG/metabolismo , Linfócitos T/metabolismo , Linfócitos T/virologia , Vacinas/genética
12.
Infect Immun ; 80(4): 1399-407, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22252874

RESUMO

Mammalian macrophage migration inhibitory factor (MIF) is a multifaceted cytokine involved in both extracellular and intracellular functions. Malaria parasites express a MIF homologue that might modulate host immune responses against blood-stage parasites, but the potential importance of MIF against other life cycle stages remains unstudied. In this study, we characterized the MIF homologue of Plasmodium yoelii throughout the life cycle, with emphasis on preerythrocytic stages. P. yoelii MIF (Py-MIF) was expressed in blood-stage parasites and detected at low levels in mosquito salivary gland sporozoites. MIF expression was strong throughout liver-stage development and localized to the cytoplasm of the parasite, with no evidence of release into the host hepatocyte. To examine the importance of Py-MIF for liver-stage development, we generated a Py-mif knockout parasite (P. yoelii Δmif). P. yoelii Δmif parasites grew normally as asexual erythrocytic-stage parasites and showed normal infection of mosquitoes. In contrast, the P. yoelii Δmif strain was attenuated during the liver stage. Mice infected with P. yoelii Δmif sporozoites either did not develop blood-stage parasitemia or exhibited a delay in the onset of blood-stage patency. Furthermore, P. yoelii Δmif parasites exhibited growth retardation in vivo. Combined, the data indicate that Plasmodium MIF is important for liver-stage development of P. yoelii, during which it is likely to play an intrinsic role in parasite development rather than modulating host immune responses to infection.


Assuntos
Fígado/parasitologia , Fatores Inibidores da Migração de Macrófagos/metabolismo , Malária/parasitologia , Plasmodium yoelii/patogenicidade , Proteínas de Protozoários/metabolismo , Animais , Anopheles/parasitologia , Linhagem Celular , Feminino , Técnicas de Inativação de Genes , Células Hep G2 , Humanos , Fatores Inibidores da Migração de Macrófagos/biossíntese , Fatores Inibidores da Migração de Macrófagos/genética , Malária/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Plasmodium yoelii/genética , Plasmodium yoelii/crescimento & desenvolvimento , Proteínas de Protozoários/biossíntese , Proteínas de Protozoários/genética , Glândulas Salivares/parasitologia , Esporozoítos/crescimento & desenvolvimento
13.
Appl Microbiol Biotechnol ; 94(1): 151-61, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22170105

RESUMO

Development of a safe, effective and affordable malaria vaccine is central to global disease control efforts. One of the most highly regarded proteins for inclusion in an asexual blood stage subunit vaccine is the 19-kDa C-terminal fragment of merozoite surface protein 1 (MSP1(19)). As production of vaccine antigens in plants can potentially overcome cost and delivery hurdles, we set out to produce MSP1(19) in plants, characterise the protein and test its immunogenicity using a mouse model. Plasmodium yoelii MSP1(19) (PyMSP1(19)) was produced in Nicotiana benthamiana using the MagnICON® deconstructed TMV-based viral vector. PyMSP1(19) yield of at least 23% total soluble protein (TSP;3-4 mg/g Fwt) were achieved using a codon-optimised construct that was targeted to the apoplast. Freeze-dried leaf powder contained at least 20 mg PyMSP1(19) per gram dry weight and the protein retained immunogenicity in this form for more than 2 years. Characterisation studies, including SDS-PAGE, mass spectrometry and circular dichroism, indicated that the plant-expressed PyMSP1(19) was similar to its Escherichia coli- and Saccharomyces cerevisiae-expressed counterparts. Purified plant-made PyMSP1(19) induced strong immune responses following intraperitoneal immunisation, although titres were lower than those induced by an equivalent dose of purified E. coli-expressed PyMSP1(19). The reason for this is uncertain but may be due to differences in the oligomerisation profile of the vaccines. The plant-made PyMSP1(19) vaccine was also found to be orally immunogenic when delivered alone or following immunisation with a PyMSP1(19) DNA vaccine. This study adds to an increasing body of research supporting the feasibility of plants as both a factory for the production of malaria antigens, and as a safe and affordable platform for oral delivery of a temperature-stable malaria vaccine.


Assuntos
Antígenos de Protozoários/genética , Antígenos de Protozoários/imunologia , Malária/imunologia , Proteína 1 de Superfície de Merozoito/genética , Proteína 1 de Superfície de Merozoito/imunologia , Nicotiana/genética , Plasmodium yoelii/imunologia , Motivos de Aminoácidos , Animais , Antígenos de Protozoários/química , Feminino , Expressão Gênica , Humanos , Imunização , Malária/parasitologia , Malária/prevenção & controle , Vacinas Antimaláricas/administração & dosagem , Vacinas Antimaláricas/química , Vacinas Antimaláricas/genética , Vacinas Antimaláricas/imunologia , Proteína 1 de Superfície de Merozoito/química , Camundongos , Camundongos Endogâmicos BALB C , Plasmodium yoelii/química , Plasmodium yoelii/genética , Plasmodium yoelii/crescimento & desenvolvimento , Nicotiana/metabolismo
14.
Cell Host Microbe ; 9(6): 451-62, 2011 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-21669394

RESUMO

While subunit vaccines have shown partial efficacy in clinical trials, radiation-attenuated sporozoites (RAS) remain the "gold standard" for sterilizing protection against Plasmodium infection in human vaccinees. The variability in immunogenicity and replication introduced by the extensive, random DNA damage necessary to generate RAS could be overcome by genetically attenuated parasites (GAP) designed via gene deletion to arrest at defined points during liver-stage development. Here, we demonstrate the principle that late liver stage-arresting GAP induce larger and broader CD8 T cell responses that provide superior protection in inbred and outbred mice compared to RAS or early-arresting GAP immunizations. Late liver stage-arresting GAP also engender high levels of cross-stage and cross-species protection and complete protection when administered by translationally relevant intradermal or subcutaneous routes. Collectively, our results underscore the potential utility of late liver stage-arresting GAP as broadly protective next-generation live-attenuated malaria vaccines and support their potential as a powerful model for identifying antigens to generate cross-stage protection.


Assuntos
Fígado/parasitologia , Vacinas Antimaláricas/imunologia , Malária/imunologia , Plasmodium berghei/genética , Plasmodium berghei/imunologia , Plasmodium yoelii/genética , Plasmodium yoelii/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/parasitologia , Deleção de Genes , Humanos , Fígado/imunologia , Malária/parasitologia , Malária/prevenção & controle , Vacinas Antimaláricas/administração & dosagem , Vacinas Antimaláricas/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Plasmodium berghei/crescimento & desenvolvimento , Plasmodium yoelii/crescimento & desenvolvimento , Esporozoítos/crescimento & desenvolvimento , Esporozoítos/imunologia , Vacinação , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia
15.
FEMS Microbiol Lett ; 318(2): 152-8, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21366672

RESUMO

Invasion of the erythrocyte by the invasive form of the malaria parasite, the merozoite, is a complex process involving numerous parasite proteins. The reticulocyte-binding protein homologues (RH) family of merozoite proteins has been previously shown to play an important role in the invasion process. Previously, it has been shown that the RH proteins of Plasmodium yoelii, Py235, play a role as an ATP/ADP sensor. Binding of Py235 to the erythrocyte surface is increased in the presence of ATP, while ADP has an inhibitory effect. The sensor domain of Py235 is called NBD94 and the segment that has been shown to covalently bind the adenine nucleotide is made up by the residues (483) FNEIKEKLKHYNFDDFVKEE(502) . Here, we report on the solution nuclear magnetic resonance structure of this peptide (NBD94(483-502) ) showing the presence of an α-helix between amino acid residues 485 and 491. The N- and C-terminal segments of the structure bend at tyrosine 493, a residue important for ATP binding. Importantly, erythrocyte-binding assays demonstrate that NBD94(483-502) can directly interfere with the binding of native Py235 to erythrocytes, suggesting a direct role of this region in erythrocyte binding. The data will provide the foundation for future studies to identify new compounds that directly interfere with the invasion process.


Assuntos
Eritrócitos/parasitologia , Malária/parasitologia , Proteínas de Membrana/química , Plasmodium yoelii/química , Proteínas de Protozoários/química , Difosfato de Adenosina/metabolismo , Trifosfato de Adenosina/metabolismo , Sequência de Aminoácidos , Animais , Eritrócitos/metabolismo , Humanos , Espectroscopia de Ressonância Magnética , Malária/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Plasmodium yoelii/genética , Plasmodium yoelii/metabolismo , Ligação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo
16.
Infect Immun ; 78(5): 2182-8, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20194600

RESUMO

Immunization with live Plasmodium sporozoites under chloroquine prophylaxis (Spz plus CQ) induces sterile immunity against sporozoite challenge in rodents and, more importantly, in humans. Full protection is obtained with substantially fewer parasites than with the classic immunization with radiation-attenuated sporozoites. The sterile protection observed comprised a massive reduction in the hepatic parasite load and an additional effect at the blood stage level. Differences in the immune responses induced by the two protocols occur but are as yet little characterized. We have previously demonstrated that in mice immunized with irradiated sporozoites, immune responses against the circumsporozoite protein (CSP), the major component of the sporozoite's surface and the leading malaria vaccine candidate, were not essential for sterile protection. Here, we have employed transgenic Plasmodium berghei parasites in which the endogenous CSP was replaced by that of Plasmodium yoelii, another rodent malaria species, to assess the role of CSP in the sterile protection induced by the Spz-plus-CQ protocol. The data demonstrated that this role was minor because sterile immunity was obtained irrespective of the origin of CSP expressed by the parasites in this model of protection. The immunity was obtained through a single transient exposure of the host to the immunizing parasites (preerythrocytic and erythrocytic), a dose much smaller than that required for immunization with radiation-attenuated sporozoites.


Assuntos
Vacinas Antimaláricas/imunologia , Malária/prevenção & controle , Plasmodium berghei/imunologia , Plasmodium yoelii/imunologia , Proteínas de Protozoários/imunologia , Animais , Feminino , Interferon gama/metabolismo , Leucócitos Mononucleares/imunologia , Fígado/parasitologia , Camundongos , Camundongos Endogâmicos BALB C , Plasmodium berghei/genética , Plasmodium yoelii/genética , Baço/imunologia , Esporozoítos/imunologia
17.
Exp Parasitol ; 126(3): 381-8, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20193682

RESUMO

Pyrimethamine is an antimalarial drug that has also been used successfully to treat autoimmune diseases such as lymphoproliferative syndrome. In this work, the effect of pyrimethamine (PYR) on the production of free radicals in malaria-infected mice was studied to better understand the drug's immunomodulatory properties. BALB/c and CBA/Ca mice were infected with Plasmodium yoelii 17XL. Seven days after infection, mice were treated with PYR or vehicle and sacrificed 24h later. Treatment with PYR increased superoxide dismutase and glutathione peroxidase activities in erythrocytes and the liver, augmented the levels of nitric oxide in the serum, and upregulated mRNA levels of superoxide dismutase, glutathione peroxidase, catalase, and iNOS in the spleen. In addition, PYR increased lipoperoxidation and protein carbonylation in infected mice. Our results indicate that P. yoelii 17XL reduces oxidative stress in infected cells, while PYR induces it, which is associated with increased parasite elimination. Thus, it is possible that oxidative stress generated by pyrimethamine is also involved in its immunomodulatory mechanism of action.


Assuntos
Antimaláricos/farmacologia , Malária/tratamento farmacológico , Estresse Oxidativo/efeitos dos fármacos , Plasmodium yoelii/efeitos dos fármacos , Pirimetamina/farmacologia , Animais , Antimaláricos/uso terapêutico , Catalase/biossíntese , Catalase/efeitos dos fármacos , Catalase/genética , Eritrócitos/efeitos dos fármacos , Eritrócitos/metabolismo , Feminino , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Glutationa Peroxidase/biossíntese , Glutationa Peroxidase/efeitos dos fármacos , Glutationa Peroxidase/genética , Peroxidação de Lipídeos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Malária/imunologia , Malária/metabolismo , Masculino , Malondialdeído/análise , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos CBA , Óxido Nítrico/sangue , Óxido Nítrico Sintase Tipo II/biossíntese , Óxido Nítrico Sintase Tipo II/efeitos dos fármacos , Óxido Nítrico Sintase Tipo II/genética , Estresse Oxidativo/imunologia , Plasmodium yoelii/genética , Plasmodium yoelii/imunologia , Carbonilação Proteica/efeitos dos fármacos , Pirimetamina/uso terapêutico , Organismos Livres de Patógenos Específicos , Baço/efeitos dos fármacos , Baço/metabolismo , Superóxido Dismutase/biossíntese , Superóxido Dismutase/efeitos dos fármacos , Superóxido Dismutase/genética , Fatores de Tempo
18.
J Immunol ; 183(9): 5870-8, 2009 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-19812194

RESUMO

The production of IFN-gamma by CD8(+) T cells is an important hallmark of protective immunity induced by irradiation-attenuated sporozoites against malaria. Here, we demonstrate that protracted sterile protection conferred by a Plasmodium yoelii genetically attenuated parasite (PyGAP) vaccine was completely dependent on CD8(+) T lymphocytes but only partially dependent on IFN-gamma. We used live cell imaging to document that CD8(+) CTL from PyGAP-immunized mice directly killed hepatocyte infected with a liver stage parasite. Immunization studies with perforin and IFN-gamma knockout mice also indicated that the protection was largely dependent on perforin-mediated effector mechanisms rather than on IFN-gamma. This was further supported by our observation that both liver and spleen CD8(+) T cells from PyGAP-immunized mice induced massive apoptosis of liver stage-infected hepatocytes in vitro without the release of detectable IFN-gamma and TNF-alpha. Conversely, CD8(+) T cells isolated from naive mice that had survived wild-type P. yoelii sporozoite infection targeted mainly sporozoite-traversed and uninfected hepatocytes, revealing an immune evasion strategy that might be used by wild-type parasites to subvert host immune responses during natural infection. However, CTLs from wild-type sporozoite-challenged mice could recognize and kill infected hepatocytes that were pulsed with circumsporozoite protein. Additionally, protection in PyGAP-immunized mice directly correlated with the magnitude of effector memory CD8(+) T cells. Our findings implicate CTLs as key immune effectors in a highly protective PyGAP vaccine for malaria and emphasize the critical need to define surrogate markers for correlates of protection, apart from IFN-gamma.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Comunicação Celular/imunologia , Citotoxicidade Imunológica/genética , Hepatócitos/imunologia , Hepatócitos/parasitologia , Hepatopatias Parasitárias/prevenção & controle , Vacinas Antimaláricas/genética , Plasmodium yoelii/genética , Animais , Linfócitos T CD8-Positivos/parasitologia , Linfócitos T CD8-Positivos/patologia , Comunicação Celular/genética , Feminino , Hepatócitos/patologia , Memória Imunológica/genética , Interferon gama/deficiência , Interferon gama/genética , Hepatopatias Parasitárias/imunologia , Hepatopatias Parasitárias/parasitologia , Malária/imunologia , Malária/parasitologia , Malária/prevenção & controle , Vacinas Antimaláricas/administração & dosagem , Vacinas Antimaláricas/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos Transgênicos , Perforina/deficiência , Perforina/genética , Plasmodium yoelii/imunologia , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia
19.
Infect Immun ; 77(5): 1798-806, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19237530

RESUMO

Immunization of mice with Plasmodium berghei or Plasmodium yoelii synthetic linear peptide chimeras (LPCs) based on the circumsporozoite protein protects against experimental challenge with viable sporozoites. The immunogenicity of LPCs is significantly enhanced by spontaneous polymerization. To better understand the antigenic properties of polymeric antimalarial peptides, we studied the immune responses elicited in mice immunized with a polymer or a monomer of a linear peptide construct specific for P. yoelii and compared the responses of antigen-presenting cells following incubation with both peptide species. Efficient uptake of the polymeric peptide in vitro resulted in higher expression of the coactivation markers CD80, CD40, and CD70 on dendritic cells and higher proinflammatory cytokine production than with the monomeric peptide. Macropinocytosis seems to be the main route used by polymeric peptides internalized by antigen-presenting cells. Spontaneous polymerization of synthetic antimalarial-peptide constructs to target professional antigen-presenting cells shows promise for simple delivery of subunit malaria vaccines.


Assuntos
Vacinas Antimaláricas/genética , Vacinas Antimaláricas/imunologia , Plasmodium berghei/imunologia , Plasmodium yoelii/imunologia , Multimerização Proteica/imunologia , Proteínas de Protozoários/imunologia , Animais , Células Apresentadoras de Antígenos/imunologia , Antígeno B7-1/biossíntese , Ligante CD27/biossíntese , Antígenos CD40/biossíntese , Linhagem Celular , Células Cultivadas , Citocinas/metabolismo , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Pinocitose , Plasmodium berghei/genética , Plasmodium yoelii/genética , Proteínas de Protozoários/genética , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia
20.
Artigo em Chinês | MEDLINE | ID: mdl-20232624

RESUMO

OBJECTIVE: To investigate on the effect of Plasmodium yoelii (BY265 strain) circumsporozoite protein (CSP) on the activation of nuclear transcription factor KB (NF-KB) in hepatoma cells (HepG2) stimulated by TNF-alpha. METHODS: Entire coding sequence of CSP was reverse transcribed and amplified by RT-PCR with sporozoite total RNA as template, then cloned into pFLAG-CMV8 for construction of the recombinant plasmid pFLAG-CMV8-CSP. Indirect immunofluorescence staining with rabbit anti-CSP was applied to verify the expression and distribution of FLAG-CSP fusion protein in HepG2. Three groups were established for the experiment: group A with HepG2 transfected by pFLAG-CMV8 as negative control, group B with HepG2 transfected by pFLAG-CMV8 and stimulated by 100 ng/ml TNF-alpha, and group C with HepG2 transfected by pFLAG-CMV8-CSP and stimulated by 100 ng/ml TNF-alpha. Dual-luciferase assay and EMSA were performed to detect the nuclear translocation and activation of NF-kappaB, to observe if pFLAG-CMV8-CSP suppressed the activation of NF-KB in HepG2 stimulated by TNF-alpha. RESULT: The expression of pFLAG-CMV8-CSP was localized on cytoplasm of HepG2. The activity ratio of firefly luciferase to Renilla luciferase in group C (0.228 +/- 0.029) was significantly lower than both groups A (0.438 +/- 0.085) and B (0.571 +/- 0.030) (P < 0.05). EMSA showed that the band in group C was significantly weaker than group B. CONCLUSION: Plasmodium yoelii CSP localizes in the cytoplasm of HepG2 and inhibits the activation and nuclear translocation of NF-kappaB in HepG2 stimulated by TNF-alpha.


Assuntos
NF-kappa B/metabolismo , Plasmodium yoelii/genética , Proteínas de Protozoários/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Animais , Células Hep G2 , Humanos , Plasmídeos , RNA de Protozoário , Esporos de Protozoários/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA