Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Sci Rep ; 8(1): 1118, 2018 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-29348479

RESUMO

Vaccine development against Plasmodium vivax malaria lags behind that for Plasmodium falciparum. To narrow this gap, we administered recombinant antigens based on P. vivax circumsporozoite protein (CSP) to mice. We expressed in Pichia pastoris two chimeric proteins by merging the three central repeat regions of different CSP alleles (VK210, VK247, and P. vivax-like). The first construct (yPvCSP-AllFL) contained the fused repeat regions flanked by N- and C-terminal regions. The second construct (yPvCSP-AllCT) contained the fused repeat regions and the C-terminal domain, plus RI region. Mice were vaccinated with three doses of yPvCSP in adjuvants Poly (I:C) or Montanide ISA720. We also used replication-defective adenovirus vectors expressing CSP of human serotype 5 (AdHu5) and chimpanzee serotype 68 (AdC68) for priming mice which were subsequently boosted twice with yPvCSP proteins in Poly (I:C) adjuvant. Regardless of the regime used, immunized mice generated high IgG titres specific to all CSP alleles. After challenge with P. berghei ANKA transgenic parasites expressing Pb/PvVK210 or Pb/PvVK247 sporozoites, significant time delays for parasitemia were observed in all vaccinated mice. These vaccine formulations should be clinically tried for their potential as protective universal vaccine against P. vivax malaria.


Assuntos
Vacinas Antimaláricas/imunologia , Malária Vivax/imunologia , Malária Vivax/prevenção & controle , Plasmodium vivax/imunologia , Proteínas de Protozoários/imunologia , Proteínas Recombinantes/imunologia , Adenoviridae/genética , Sequência de Aminoácidos , Animais , Anticorpos Antiprotozoários/imunologia , Afinidade de Anticorpos/imunologia , Modelos Animais de Doenças , Feminino , Vetores Genéticos/administração & dosagem , Vetores Genéticos/química , Imunização , Imunogenicidade da Vacina , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Vacinas Antimaláricas/genética , Malária Vivax/mortalidade , Camundongos , Plasmodium vivax/genética , Proteínas de Protozoários/química , Proteínas de Protozoários/genética
2.
Front Immunol ; 8: 1275, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29075260

RESUMO

Plasmodium vivax is the most common species that cause malaria outside of the African continent. The development of an efficacious vaccine would contribute greatly to control malaria. Recently, using bacterial and adenoviral recombinant proteins based on the P. vivax circumsporozoite protein (CSP), we demonstrated the possibility of eliciting strong antibody-mediated immune responses to each of the three allelic forms of P. vivax CSP (PvCSP). In the present study, recombinant proteins representing the PvCSP alleles (VK210, VK247, and P. vivax-like), as well as a hybrid polypeptide, named PvCSP-All epitopes, were generated. This hybrid containing the conserved C-terminal of the PvCSP and the three variant repeat domains in tandem were successfully produced in the yeast Pichia pastoris. After purification and biochemical characterization, they were used for the experimental immunization of C57BL/6 mice in a vaccine formulation containing the adjuvant Poly(I:C). Immunization with a recombinant protein expressing all three different allelic forms in fusion elicited high IgG antibody titers reacting with all three different allelic variants of PvCSP. The antibodies targeted both the C-terminal and repeat domains of PvCSP and recognized the native protein on the surface of P. vivax sporozoites. More importantly, mice that received the vaccine formulation were protected after challenge with chimeric Plasmodium berghei sporozoites expressing CSP repeats of P. vivax sporozoites (Pb/PvVK210). Our results suggest that it is possible to elicit protective immunity against one of the most common PvCSP alleles using soluble recombinant proteins expressed by P. pastoris. These recombinant proteins are promising candidates for clinical trials aiming to develop a multiallele vaccine against P. vivax malaria.

3.
Vaccine ; 34(38): 4501-4506, 2016 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-27502569

RESUMO

A number of studies have shown that CD8+ T cells mediate protective anti-malaria immunity in a mouse model. However, whether human CD8+ T cells play a role in protection against malaria remains unknown. We recently established human immune system (HIS) mice harboring functional human CD8+ T cells (HIS-CD8 mice) by transduction with HLA-A∗0201 and certain human cytokines using recombinant adeno-associated virus-based gene transfer technologies. These HIS-CD8 mice mount a potent, antigen-specific HLA-A∗0201-restricted human CD8+ T-cell response upon immunization with a recombinant adenovirus expressing a human malaria antigen, the Plasmodium falciparum circumsporozoite protein (PfCSP), termed AdPfCSP. In the present study, we challenged AdPfCSP-immunized HIS-CD8 mice with transgenic Plasmodium berghei sporozoites expressing full-length PfCSP and found that AdPfCSP-immunized (but not naïve) mice were protected against subsequent malaria challenge. The level of the HLA-A∗0201-restricted, PfCSP-specific human CD8+ T-cell response was closely correlated with the level of malaria protection. Furthermore, depletion of human CD8+ T cells from AdPfCSP-immunized HIS-CD8 mice almost completely abolished the anti-malaria immune response. Taken together, our data show that human CD8+ T cells mediate protective anti-malaria immunity in vivo.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Vacinas Antimaláricas/imunologia , Malária Falciparum/imunologia , Malária Falciparum/prevenção & controle , Animais , Dependovirus , Vetores Genéticos , Antígeno HLA-A2/genética , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos Transgênicos , Plasmodium falciparum , Proteínas de Protozoários/imunologia , Esporozoítos
4.
J Immunol Methods ; 427: 42-50, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26410104

RESUMO

In this study, we developed human immune system (HIS) mice that possess functional human CD4+ T cells and B cells, named HIS-CD4/B mice. HIS-CD4/B mice were generated by first introducing HLA class II genes, including DR1 and DR4, along with genes encoding various human cytokines and human B cell activation factor (BAFF) to NSG mice by adeno-associated virus serotype 9 (AAV9) vectors, followed by engrafting human hematopoietic stem cells (HSCs). HIS-CD4/B mice, in which the reconstitution of human CD4+ T and B cells resembles to that of humans, produced a significant level of human IgG against Plasmodium falciparum circumsporozoite (PfCS) protein upon immunization. CD4+ T cells in HIS-CD4/B mice, which possess central and effector memory phenotypes like those in humans, are functional, since PfCS protein-specific human CD4+ T cells secreting IFN-γ and IL-2 were detected in immunized HIS-CD4/B mice. Lastly, PfCS protein-immunized HIS-CD4/B mice were protected from in vivo challenge with transgenic P. berghei sporozoites expressing the PfCS protein. The immune sera collected from protected HIS-CD4/B mice reacted against transgenic P. berghei sporozoites expressing the PfCS protein and also inhibited the parasite invasion into hepatocytes in vitro. Taken together, these studies show that our HIS-CD4/B mice could mount protective human anti-malaria immunity, consisting of human IgG and human CD4+ T cell responses both specific for a human malaria antigen.


Assuntos
Modelos Animais de Doenças , Imunidade Humoral/imunologia , Malária Falciparum/imunologia , Camundongos Transgênicos/imunologia , Animais , Ensaio de Imunoadsorção Enzimática , Xenoenxertos , Antígenos de Histocompatibilidade Classe II , Humanos , Vacinas Antimaláricas , Camundongos , Proteínas de Protozoários/imunologia
5.
Infect Immun ; 82(2): 793-807, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24478093

RESUMO

Plasmodium vivax is the most widespread and the second most prevalent malaria-causing species in the world. Current measures used to control the transmission of this disease would benefit from the development of an efficacious vaccine. In the case of the deadly parasite P. falciparum, the recombinant RTS,S vaccine containing the circumsporozoite antigen (CSP) consistently protects 30 to 50% of human volunteers against infection and is undergoing phase III clinical trials in Africa with similar efficacy. These findings encouraged us to develop a P. vivax vaccine containing the three circulating allelic forms of P. vivax CSP. Toward this goal, we generated three recombinant bacterial proteins representing the CSP alleles, as well as a hybrid polypeptide called PvCSP-All-CSP-epitopes. This hybrid contains the conserved N and C termini of P. vivax CSP and the three variant repeat domains in tandem. We also generated simian and human recombinant replication-defective adenovirus vectors expressing PvCSP-All-CSP-epitopes. Mice immunized with the mixture of recombinant proteins in a formulation containing the adjuvant poly(I·C) developed high and long-lasting serum IgG titers comparable to those elicited by proteins emulsified in complete Freund's adjuvant. Antibody titers were similar in mice immunized with homologous (protein-protein) and heterologous (adenovirus-protein) vaccine regimens. The antibodies recognized the three allelic forms of CSP, reacted to the repeated and nonrepeated regions of CSP, and recognized sporozoites expressing the alleles VK210 and VK247. The vaccine formulations described in this work should be useful for the further development of an anti-P. vivax vaccine.


Assuntos
Vacinas Antimaláricas/imunologia , Malária Vivax/prevenção & controle , Plasmodium vivax/imunologia , Proteínas de Protozoários/imunologia , Vacinação/métodos , Adjuvantes Imunológicos/administração & dosagem , Animais , Anticorpos Antiprotozoários/sangue , Feminino , Imunoglobulina G/sangue , Vacinas Antimaláricas/administração & dosagem , Vacinas Antimaláricas/genética , Malária Vivax/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Plasmodium vivax/genética , Poli I-C/administração & dosagem , Proteínas de Protozoários/genética , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia
6.
Biomaterials ; 33(27): 6476-84, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22695068

RESUMO

Biomaterials that modulate innate and adaptive immune responses are receiving increasing interest as adjuvants for eliciting protective immunity against a variety of diseases. Previous results have indicated that self-assembling ß-sheet peptides, when fused with short peptide epitopes, can act as effective adjuvants and elicit robust and long-lived antibody responses. Here we investigated the mechanism of immunogenicity and the quality of antibody responses raised by a peptide epitope from Plasmodium falciparum circumsporozoite (CS) protein, (NANP)(3),conjugated to the self-assembling peptide domain Q11. The mechanism of adjuvant action was investigated in knockout mice with impaired MyD88, NALP3, TLR-2, or TLR-5 function, and the quality of antibodies raised against (NANP)(3)-Q11 was assessed using a transgenic sporozoite neutralizing (TSN) assay for malaria infection. (NANP)(3)-Q11 self-assembled into nanofibers, and antibody responses lasted up to 40 weeks in C57BL/6 mice. The antibody responses were T cell- and MyD88-dependent. Sera from mice primed with either irradiated sporozoites or a synthetic peptide, (T1BT*)(4)-P3C, and boosted with (NANP)(3)-Q11 showed significant increases in antibody titers and significant inhibition of sporozoite infection in TSN assays. In addition, two different epitopes could be self-assembled together without compromising the strength or duration of the antibody responses raised against either of them, making these materials promising platforms for self-adjuvanting multi-antigenic immunotherapies.


Assuntos
Formação de Anticorpos/imunologia , Epitopos/imunologia , Malária/imunologia , Nanofibras/química , Peptídeos/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Antiprotozoários/imunologia , Ensaio de Imunoadsorção Enzimática , Malária/parasitologia , Malária/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dados de Sequência Molecular , Fator 88 de Diferenciação Mieloide/metabolismo , Nanofibras/ultraestrutura , Oligopeptídeos/química , Oligopeptídeos/imunologia , Peptídeos/química , Estrutura Secundária de Proteína , Esporozoítos/imunologia , Linfócitos T/imunologia
7.
Proc Natl Acad Sci U S A ; 109(10): 3956-61, 2012 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-22355110

RESUMO

In response to environmental stresses, the mammalian serine threonine kinases PERK, GCN2, HRI, and PKR phosphorylate the regulatory serine 51 of the eukaryotic translation initiation factor 2α (eIF2α) to inhibit global protein synthesis. Plasmodium, the protozoan that causes malaria, expresses three eIF2α kinases: IK1, IK2, and PK4. Like GCN2, IK1 regulates stress response to amino acid starvation. IK2 inhibits development of malaria sporozoites present in the mosquito salivary glands. Here we show that the phosphorylation by PK4 of the regulatory serine 59 of Plasmodium eIF2α is essential for the completion of the parasite's erythrocytic cycle that causes disease in humans. PK4 activity leads to the arrest of global protein synthesis in schizonts, where ontogeny of daughter merozoites takes place, and in gametocytes that infect Anopheles mosquitoes. The implication of these findings is that drugs that reduce PK4 activity should alleviate disease and inhibit malaria transmission.


Assuntos
Plasmodium falciparum/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , eIF-2 Quinase/metabolismo , Animais , Anopheles , Códon , DNA/genética , Proteínas Fúngicas/química , Células Hep G2 , Humanos , Malária/parasitologia , Camundongos , Camundongos Endogâmicos C57BL , Modelos Genéticos , Mutação , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Serina/química
8.
Mem Inst Oswaldo Cruz ; 106 Suppl 1: 167-71, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21881771

RESUMO

Recently, we described the improved immunogenicity of new malaria vaccine candidates based on the expression of fusion proteins containing immunodominant epitopes of merozoites and Salmonella enterica serovar Typhimurium flagellin (FliC) protein as an innate immune agonist. Here, we tested whether a similar strategy, based on an immunodominant B-cell epitope from malaria sporozoites, could also generate immunogenic fusion polypeptides. A recombinant His6-tagged FliC protein containing the C-terminal repeat regions of the VK210 variant of Plasmodium vivax circumsporozoite (CS) protein was constructed. This recombinant protein was successfully expressed in Escherichia coli as soluble protein and was purified by affinity to Ni-agarose beads followed by ion exchange chromatography. A monoclonal antibody specific for the CS protein of P. vivax sporozoites (VK210) was able to recognise the purified protein. C57BL/6 mice subcutaneously immunised with the recombinant fusion protein in the absence of any conventional adjuvant developed protein-specific systemic antibody responses. However, in mice genetically deficient in expression of TLR5, this immune response was extremely low. These results extend our previous observations concerning the immunogenicity of these recombinant fusion proteins and provide evidence that the main mechanism responsible for this immune activation involves interactions with TLR5, which has not previously been demonstrated for any recombinant FliC fusion protein.


Assuntos
Flagelina/imunologia , Epitopos Imunodominantes/imunologia , Vacinas Antimaláricas/imunologia , Malária Vivax/prevenção & controle , Plasmodium falciparum/imunologia , Proteínas Recombinantes de Fusão/imunologia , Salmonella typhimurium/imunologia , Animais , Anticorpos Antiprotozoários/imunologia , Epitopos de Linfócito B/imunologia , Epitopos de Linfócito B/metabolismo , Proteínas de Escherichia coli/imunologia , Flagelina/metabolismo , Epitopos Imunodominantes/metabolismo , Vacinas Antimaláricas/metabolismo , Malária Vivax/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas de Protozoários/imunologia , Proteínas de Protozoários/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Salmonella typhimurium/metabolismo , Receptor 5 Toll-Like/imunologia
9.
Vaccine ; 29(43): 7335-42, 2011 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-21807053

RESUMO

Immunization of BALB/c mice with irradiated sporozoites (IrSp) of Plasmodium yoelii can lead to sterile immunity. The circumsporozoite protein (CSP) plays a dominant role in protection. Nevertheless after hyper-immunization with IrSp, complete protection is obtained in CSP-transgenic BALB/c mice that are T-cell tolerant to the CSP and cannot produce antibodies [CSP-Tg/JhT(-/-)]. This protection is mediated exclusively by CD8(+) T cells [1]. To identify the non-CSP protective T cell antigens, we studied the properties of 34 P. yoelii sporozoite antigens that are predicted to be secreted and to contain strong Kd-restricted CD8(+) T cell epitopes. The synthetic peptides corresponding to the epitopes were used to screen for the presence of peptide-specific CD8(+) T cells secreting interferon-γ (IFN-γ) in splenocytes from CSP-Tg/JhT(-/-) BALB/c mice hyper immunized with IrSp. However, the numbers of IFN-γ-secreting splenocytes specific for the non-CSP antigen-derived peptides were 20-100 times lower than those specific for the CSP-specific peptide. When mice were immunized with recombinant adenoviruses expressing selected non-CSP antigens, the animals were not protected against challenge with P. yoelii sporozoites although large numbers of CD8(+) specific T cells were generated.


Assuntos
Antígenos de Protozoários/imunologia , Epitopos de Linfócito T/imunologia , Vacinas Antimaláricas/imunologia , Plasmodium yoelii/imunologia , Proteínas de Protozoários/imunologia , Esporozoítos/imunologia , Adenoviridae/genética , Adenoviridae/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Feminino , Imunização , Interferon gama , Malária/imunologia , Malária/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Esporozoítos/efeitos da radiação
10.
Mem. Inst. Oswaldo Cruz ; 106(supl.1): 167-171, Aug. 2011. ilus, graf
Artigo em Inglês | LILACS | ID: lil-597258

RESUMO

Recently, we described the improved immunogenicity of new malaria vaccine candidates based on the expression of fusion proteins containing immunodominant epitopes of merozoites and Salmonella enterica serovar Typhimurium flagellin (FliC) protein as an innate immune agonist. Here, we tested whether a similar strategy, based on an immunodominant B-cell epitope from malaria sporozoites, could also generate immunogenic fusion polypeptides. A recombinant His6-tagged FliC protein containing the C-terminal repeat regions of the VK210 variant of Plasmodium vivax circumsporozoite (CS) protein was constructed. This recombinant protein was successfully expressed in Escherichia coli as soluble protein and was purified by affinity to Ni-agarose beads followed by ion exchange chromatography. A monoclonal antibody specific for the CS protein of P. vivax sporozoites (VK210) was able to recognise the purified protein. C57BL/6 mice subcutaneously immunised with the recombinant fusion protein in the absence of any conventional adjuvant developed protein-specific systemic antibody responses. However, in mice genetically deficient in expression of TLR5, this immune response was extremely low. These results extend our previous observations concerning the immunogenicity of these recombinant fusion proteins and provide evidence that the main mechanism responsible for this immune activation involves interactions with TLR5, which has not previously been demonstrated for any recombinant FliC fusion protein.


Assuntos
Animais , Camundongos , Flagelina/imunologia , Epitopos Imunodominantes/imunologia , Vacinas Antimaláricas/imunologia , Malária Vivax , Plasmodium falciparum/imunologia , Proteínas Recombinantes de Fusão/imunologia , Salmonella typhimurium/imunologia , Anticorpos Antiprotozoários/imunologia , Epitopos de Linfócito B/imunologia , Epitopos de Linfócito B , Proteínas de Escherichia coli/imunologia , Flagelina , Epitopos Imunodominantes , Vacinas Antimaláricas , Malária Vivax/imunologia , Proteínas de Protozoários/imunologia , Proteínas de Protozoários , Proteínas Recombinantes de Fusão , Salmonella typhimurium , /imunologia
11.
Antimicrob Agents Chemother ; 54(7): 2987-93, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20457823

RESUMO

Nitrogen-containing bisphosphonates, drugs used to treat bone resorption diseases, also have activity against a broad range of protists, including blood-stage Plasmodium spp. Here, we show that new-generation "lipophilic" bisphosphonates designed as anticancer agents that block protein prenylation also have potent activity against Plasmodium liver stages, with a high (>100) therapeutic index. Treatment of mice with the bisphosphonate BPH-715 and challenge with Plasmodium berghei sporozoites revealed complete protection (no blood-stage parasites after 28 days). There was also activity against blood-stage forms in vitro and a 4-day delay in the prepatent period in vivo. The lipophilic bisphosphonates have activity against a Plasmodium geranylgeranyl diphosphate synthase (GGPPS), as well as low nM activity against human farnesyl and geranylgeranyl diphosphate synthases. The most active inhibitor in vitro and in vivo had enzyme inhibitory activity similar to that of the other, less active compounds but was more lipophilic. Lipophilic bisphosphonates are thus promising leads for novel antimalarials that target liver-stage infection.


Assuntos
Antimaláricos/uso terapêutico , Difosfonatos/uso terapêutico , Fígado/fisiopatologia , Plasmodium berghei/efeitos dos fármacos , Animais , Difosfonatos/química , Células Hep G2 , Humanos , Camundongos , Modelos Biológicos , Estrutura Molecular , Plasmodium berghei/crescimento & desenvolvimento , Plasmodium berghei/patogenicidade
12.
Proc Natl Acad Sci U S A ; 107(3): 1172-7, 2010 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-20080619

RESUMO

There is yet no licensed vaccine against malaria, a serious human disease affecting mostly children, with an annual death rate of about one million. Plasmodia, the malaria-causing parasites, have two obligatory hosts: mammals or birds, in which they multiply asexually, and mosquitoes with sexual multiplication. The most common and serious type of malaria is caused by Plasmodium falciparum. The circumsporozoite protein (CSP), a major surface antigen of sporozoites, is a protective antigen. A unique feature of P. falciparum CSP is its large central domain composed of over 30 tetrapeptide repeats of Asn-Ala-Asn-Pro (NANP). Several NANP peptide-protein conjugates were tested clinically but elicited a low level of CSP antibodies for a short duration. To provide a CSP-based candidate vaccine, we investigated recombinant CSP and NANP conjugates of various peptide lengths, with different N-terminal amino acids, bound at different ratios to various carrier proteins. Injected into mice, CSP alone and CSP or NANP conjugates induced antibodies with booster responses and were positive by the sporozoite immunofluorescent assay. The use of the mosquito stage P. falciparum ookinete surface protein, Pfs25, cross-linked onto itself as a carrier for NANP, induced in mice high levels of uniquely long-lasting antibodies to both vaccine components with secondary biological activities, that will provide immunity to liver infection by sporozoites and block transmission by mosquitoes.


Assuntos
Vacinas Antimaláricas/imunologia , Peptídeos/química , Plasmodium falciparum/imunologia , Proteínas de Protozoários/química , Animais , Anticorpos Antiprotozoários/imunologia , Ensaio de Imunoadsorção Enzimática , Epitopos/imunologia , Feminino , Imunofluorescência , Cromatografia Gasosa-Espectrometria de Massas , Vacinas Antimaláricas/química , Camundongos , Peptídeos/imunologia , Proteínas de Protozoários/imunologia , Proteínas Recombinantes/química , Proteínas Recombinantes/imunologia , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Linfócitos T/imunologia
13.
PLoS One ; 4(2): e4480, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19214236

RESUMO

Immunization with radiation attenuated Plasmodium sporozoites (RAS) elicits sterile protective immunity against sporozoite challenge in murine models and in humans. Similarly to RAS, the genetically attenuated sporozoites (GAPs) named uis3(-), uis4(-) and P36p(-) have arrested growth during the liver stage development, and generate a powerful protective immune response in mice. We compared the protective mechanisms in P. yoelii RAS, uis3(-) and uis4(-) in BALB/c mice. In RAS and GAPs, sterile immunity is only achieved after one or more booster injections. There were no differences in the immune responses to the circumsporozoite protein (CSP) generated by RAS and GAPs. To evaluate the role of non-CSP T-cell antigens we immunized antibody deficient, CSP-transgenic BALB/c mice, that are T cell tolerant to CSP, with P. yoelii RAS or with uis3(-) or uis4(-) GAPs, and challenged them with wild type sporozoites. In every instance the parasite liver stage burden was approximately 3 logs higher in antibody deficient CSP transgenic mice as compared to antibody deficient mice alone. We conclude that CSP is a powerful protective antigen in both RAS and GAPs viz., uis3(-) and uis4(-) and that the protective mechanisms are similar independently of the method of sporozoite attenuation.


Assuntos
Plasmodium , Esporozoítos , Animais , Antígenos de Protozoários/imunologia , Humanos , Imunização , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Plasmodium/genética , Plasmodium/imunologia , Plasmodium/efeitos da radiação , Vacinas Protozoárias/imunologia , Esporozoítos/imunologia , Esporozoítos/efeitos da radiação
14.
J Immunol Methods ; 292(1-2): 157-64, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15350520

RESUMO

The circumsporozoite (CS) protein is the major surface protein of Plasmodium sporozoites. Antibodies to the immunodominant repeat domain of CS immobilize sporozoites and prevent infection of hepatocytes. Plasmodium falciparum vaccines containing CS repeats are undergoing human trials in endemic areas, and proof of efficacy has been obtained. The correlates of protection are under investigation. Levels of anti-repeat antibodies in the serum of the human volunteers have been measured mostly by enzyme-linked immunosorbent assay (ELISA) and IFA. Assays that measure the effect of the serum antibodies on parasite infectivity (serum neutralization assays SNAs) are not usually performed because they require a susceptible host and P. falciparum sporozoites are highly infectious only to humans. To overcome this limitation, we developed a new assay named transgenic sporozoite neutralization assay (TSNA) that uses as neutralization target, a transgenic rodent malaria parasite Plasmodium berghei that bears the P. falciparum CS repeats [CS(Pf)]. Following incubation with human serum, CS(Pf) infectivity of HepG2 cells is evaluated by real-time PCR. We have compared ELISA titers and TSNAs in a limited number of sera from humans immunized with (T1B)4 MAP, a peptide vaccine containing P. falciparum CS repeats. A comparison between the two assays did not reach significance (p=0.175) when analyzed by non-parametric Spearman correlation method. Ongoing human trials of CS-based vaccines should provide an opportunity to determine whether TSNAs will provide better correlates of protective immunity than ELISA assays.


Assuntos
Testes de Neutralização , Plasmodium falciparum/imunologia , Proteínas de Protozoários/imunologia , Esporozoítos/imunologia , Animais , Linhagem Celular Tumoral , Citocalasina D/farmacologia , Ensaio de Imunoadsorção Enzimática , Humanos , Soros Imunes/imunologia , Vacinas Antimaláricas/imunologia , Reação em Cadeia da Polimerase
16.
J Cell Sci ; 116(Pt 1): 39-49, 2003 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-12456714

RESUMO

Apicomplexan host cell invasion and gliding motility depend on the parasite's actomyosin system located beneath the plasma membrane of invasive stages. Myosin A (MyoA), a class XIV unconventional myosin, is the motor protein. A model has been proposed to explain how the actomyosin motor operates but little is known about the components, topology and connectivity of the motor complex. Using the MyoA neck and tail domain as bait in a yeast two-hybrid screen we identified MTIP, a novel 24 kDa protein that interacts with MyoA. Deletion analysis shows that the 15 amino-acid C-terminal tail domain of MyoA, rather than the neck domain, specifically interacts with MTIP. In Plasmodium sporozoites MTIP localizes to the inner membrane complex (IMC), where it is found clustered with MyoA. The data support a model for apicomplexan motility and invasion in which the MyoA motor protein is associated via its tail domain with MTIP, immobilizing it at the outer IMC membrane. The head domain of the immobilized MyoA moves actin filaments that, directly or via a bridging protein, connect to the cytoplasmic domain of a transmembrane protein of the TRAP family. The actin/TRAP complex is then redistributed by the stationary MyoA from the anterior to the posterior end of the zoite, leading to its forward movement on a substrate or to penetration of a host cell.


Assuntos
Proteínas de Transporte/isolamento & purificação , Membrana Celular/metabolismo , Movimento Celular/genética , Proteínas do Citoesqueleto , Proteínas de Membrana/isolamento & purificação , Proteínas Motores Moleculares/isolamento & purificação , Plasmodium yoelii/metabolismo , Esporozoítos/metabolismo , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/metabolismo , Sequência de Aminoácidos/genética , Animais , Sequência de Bases/genética , Proteínas de Transporte/genética , Compartimento Celular/genética , Membrana Celular/ultraestrutura , DNA Complementar/análise , DNA Complementar/genética , Interações Hospedeiro-Parasita/genética , Humanos , Substâncias Macromoleculares , Proteínas de Membrana/genética , Microscopia Eletrônica , Modelos Biológicos , Proteínas Motores Moleculares/genética , Dados de Sequência Molecular , Miosina não Muscular Tipo IIA/metabolismo , Plasmodium yoelii/patogenicidade , Plasmodium yoelii/ultraestrutura , Ligação Proteica/genética , Estrutura Terciária de Proteína/genética , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Esporozoítos/patogenicidade , Esporozoítos/ultraestrutura , Células Tumorais Cultivadas , Técnicas do Sistema de Duplo-Híbrido
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA