Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 122
Filtrar
1.
Vaccine ; 37(50): 7443-7450, 2019 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-27916410

RESUMO

On 1 February 2016, in the context of the ongoing Zika virus epidemic, the WHO declared that the recently reported clusters of microcephaly and other neurological disorders constituted a Public Health Emergency of International Concern (PHEIC). In response, WHO in collaboration with UNICEF and a working group of independent subject matter experts developed a Zika virus vaccine Target Product Profile (TPP) for use in an emergency, or in a future outbreak scenario. The drafting process of the Zika virus vaccine TPP included the opportunity for public comment, as well as consultation with epidemiologists, flavivirus vaccine subject matter experts, vaccine developers and global regulators to consider the regulatory expectations and potential emergency use pathways for a vaccine with the characteristics described in the TPP. This report summarizes an expert consultation held 6-7 June 2016 on the regulatory considerations for a Zika vaccine for emergency use.


Assuntos
Surtos de Doenças/prevenção & controle , Vacinas Virais/imunologia , Infecção por Zika virus/imunologia , Zika virus/imunologia , Animais , Emergências , Humanos , Saúde Pública/métodos , Encaminhamento e Consulta , Organização Mundial da Saúde
2.
Vaccine ; 19(32): 4661-8, 2001 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-11535314

RESUMO

The gene encoding the Plasmodium yoelii homologue of P. falciparum merozoite surface proteins 4 (MSP4) and 5 (MSP5) has been expressed in Escherichia coli and Saccharomyces cerevisiae. The protein contains a single epidermal growth factor (EGF)-like domain and is expressed in a form lacking the predicted N-terminal signal and glycosyl phosphatidylinositol (GPI) attachment sequences. The recombinant protein derived from E. coli (EcMSP4/5) was highly effective at protecting mice against lethal challenge with 10(5) parasites of the P. yoelii YM strain. In contrast, the protective efficacy of yeast-derived MSP4/5 (yMSP4/5) was considerably less. The antibody titres in both groups were significantly different with mice immunised with yeast-derived protein showing significantly lower pre-challenge antibody responses. There was a significant inverse correlation between antibody levels as measured by ELISA and peak parasitaemia. Mice immunised with EcMSP4/5 produced anti-PyMSP4/5 antibodies predominantly of the IgG2a and IgG2b isotypes, whereas, mice immunised with yMSP4/5 mainly produced antibodies of the IgG1 isotype. The differences in antibody titres and subtype distribution may account for the observed differences in protective efficacy of these protein preparations. Levels of protective efficacy of MSP4/5 were compared with that obtained using P. yoelii MSP1 produced in S. cerevisiae. Levels of protection induced by E. coli derived MSP4/5 were superior to those induced by MSP1 which in turn were better than those induced by yeast-derived MSP4/5.


Assuntos
Antígenos de Protozoários/imunologia , Escherichia coli/metabolismo , Vacinas Antimaláricas/imunologia , Malária/prevenção & controle , Proteínas de Membrana/imunologia , Plasmodium yoelii/imunologia , Proteínas de Protozoários/imunologia , Saccharomyces cerevisiae/metabolismo , Animais , Anticorpos Antiprotozoários/biossíntese , Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/biossíntese , Antígenos de Protozoários/genética , Antígenos de Protozoários/isolamento & purificação , Feminino , Imunoglobulina G/biossíntese , Imunoglobulina G/imunologia , Isotipos de Imunoglobulinas/biossíntese , Isotipos de Imunoglobulinas/imunologia , Malária/imunologia , Vacinas Antimaláricas/biossíntese , Vacinas Antimaláricas/genética , Vacinas Antimaláricas/isolamento & purificação , Proteínas de Membrana/biossíntese , Proteínas de Membrana/genética , Proteínas de Membrana/isolamento & purificação , Camundongos , Camundongos Endogâmicos BALB C , Parasitemia/imunologia , Plasmodium yoelii/genética , Estrutura Terciária de Proteína , Proteínas de Protozoários/biossíntese , Proteínas de Protozoários/genética , Proteínas de Protozoários/isolamento & purificação , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/isolamento & purificação , Especificidade da Espécie , Relação Estrutura-Atividade , Vacinação
3.
Am J Trop Med Hyg ; 64(3-4): 204-6, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11442218

RESUMO

To further characterize protective-type (IgG1/IgG3) antibody responses to Plasmodium falciparum blood stage in putatively immune individuals' plasma, we have tested for various analogs of the 19 kDa C-terminus of the MSP1 antigen obtained as secreted recombinant proteins from Saccharomyces cerevisiae. One of four proteins was then identified on the basis of consistent IgG3, along with less variable IgG1 recognition. This protein has thus been selected for further functional assays of IgG1/IgG3 antibodies.


Assuntos
Imunoglobulina G/biossíntese , Vacinas Antimaláricas/imunologia , Malária Falciparum/prevenção & controle , Proteína 1 de Superfície de Merozoito/imunologia , Plasmodium falciparum/imunologia , Animais , Ensaio de Imunoadsorção Enzimática , Humanos , Malária Falciparum/epidemiologia , Proteínas Recombinantes/imunologia , Estações do Ano , Senegal/epidemiologia , Vacinas Sintéticas/imunologia
4.
Exp Parasitol ; 97(2): 61-9, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11281702

RESUMO

Gozar, M. M. G., Muratova, O., Keister, D. B., Kensil, C. R., Price, V. L., and Kaslow, D. C. 2001. Plasmodium falciparum: Immunogenicity of alum-adsorbed clinical-grade TBV25-28, a yeast-secreted malaria transmission-blocking vaccine candidate. Experimental Parasitology 97, 61-69. The fusion of Pfs25 and Pfs28, two major surface antigens on zygotes and ookinetes of Plasmodium falciparum, as a single recombinant protein (TBV25-28) was previously shown to elicit potent transmission-blocking antibodies in mice. Clinical-grade TBV25-28 was subsequently manufactured and its potency was evaluated in rabbits. Rabbits received three doses of either clinical-grade TBV25H or clinical-grade TBV25-28 adsorbed to alum with or without QS-21. As measured in a standard membrane-feeding assay, addition of QS-21 to the formulations appeared to enhance transmission-blocking potency of rabbit sera after two vaccinations but not after three vaccinations. Surprisingly, TBV25H elicited more potent transmission-blocking antibodies than did TBV25-28, a result strikingly different from those of previous mouse experiments using research-grade TBV25-28. The apparent decrease in potency of clinical-grade TBV25-28 in rabbits appears to reflect an enhancement in potency of clinical-grade TBV25H in a new formulation rather than simply a species difference in immunogenicity of TBV25-28.


Assuntos
Vacinas Antimaláricas/imunologia , Malária Falciparum/prevenção & controle , Plasmodium falciparum/imunologia , Proteínas de Protozoários/imunologia , Proteínas Recombinantes de Fusão/imunologia , Adjuvantes Imunológicos , Compostos de Alúmen , Animais , Anticorpos Antiprotozoários/biossíntese , Anticorpos Antiprotozoários/sangue , Ensaio de Imunoadsorção Enzimática , Concentração de Íons de Hidrogênio , Imunização Secundária , Vacinas Antimaláricas/normas , Camundongos , Coelhos , Saponinas , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/normas
5.
Infect Immun ; 69(3): 1536-46, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11179324

RESUMO

In an attempt to produce a more defined, clinical-grade version of a vaccine based on Plasmodium falciparum merozoite surface protein 1 (MSP1), we evaluated the efficacy of two recombinant forms of MSP1 in an Aotus nancymai challenge model system. One recombinant vaccine, bvMSP1(42), based on the 42-kDa C-terminal portion of MSP1, was expressed as a secreted protein in baculovirus-infected insect cells. A highly pure baculovirus product could be reproducibly expressed and purified at yields in excess of 8 mg of pure protein per liter of culture. This protein, when tested for efficacy in the Aotus challenge model, gave significant protection, with only one of seven monkeys requiring treatment for uncontrolled parasitemia after challenge with P. falciparum. The second recombinant protein, P30P2MSP1(19), has been used in previous studies and is based on the smaller, C-terminal 19-kDa portion of MSP1 expressed in Saccharomyces cerevisiae. Substantial changes were made in its production process to optimize expression. The optimum form of this vaccine antigen (as judged by in vitro and in vivo indicators) was then evaluated, along with bvMSP1(42), for efficacy in the A. nancymai system. The new formulation of P30P3MSP1(19) performed significantly worse than bvMSP1(42) and appeared to be less efficacious than we have found in the past, with four of seven monkeys in the vaccinated group requiring treatment for uncontrolled parasitemia. With both antigens, protection was seen only when high antibody levels were obtained by formulation of the vaccines in Freund's adjuvant. Vaccine formulation in an alternate adjuvant, MF59, resulted in significantly lower antibody titers and no protection.


Assuntos
Vacinas Antimaláricas/uso terapêutico , Malária Falciparum/prevenção & controle , Proteína 1 de Superfície de Merozoito/uso terapêutico , Plasmodium falciparum/imunologia , Vacinação , Animais , Anticorpos Antiprotozoários/sangue , Aotidae , Baculoviridae/genética , Variação Genética , Proteína 1 de Superfície de Merozoito/genética , Parasitemia , Coelhos , Proteínas Recombinantes de Fusão/uso terapêutico , Tecnologia Farmacêutica/métodos , Toxina Tetânica/uso terapêutico , Vacinas Sintéticas/uso terapêutico
6.
J Virol Methods ; 92(1): 91-7, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11164922

RESUMO

Recombinant adenoviruses are used widely in gene therapy research. Much work has been carried out to remove specific components of the wild type adenovirus (e.g. E1 gene) in order to make them safer for human use. In addition to such efforts, it is vitally important to ensure that the production of recombinant adenoviruses meet safety guidelines not only with regard to the absence of replication competent adenoviruses but for other variant species that may be present in a viral preparation. In this report, a time and cost efficient method is described for the isolation of full length adenovirus genomes without resorting to plaque purification. The procedure uses a bacterial homologous recombination system and results in the conversion of the double-stranded linear adenovirus genome into a circularized plasmid form that can be easily analyzed by restriction digestion, PCR, DNA sequencing or used in transient transfection studies. Also, the adenovirus plasmids that are generated may also be rescued back into virus form if needed. The entire procedure takes 4 days or less instead of weeks that plaque purification or dilution cloning requires. Furthermore, the method does not require the use of tissue culture materials or facilities. More importantly, this procedure allows for a more extensive and thorough examination of any viral preparation, since it allows for the detection of variants incapable of propagation without the assistance of co-infecting intact adenoviral genomes. Under standard conditions of plaque purification, these variant genomes are not detected. It is predicted that far more variant genomes will be observed using this rapid method than would otherwise be detected by standard plaque purification methods.


Assuntos
Adenoviridae/genética , Genoma Viral , Recombinação Genética , Células Clonais , Clonagem Molecular , DNA Viral/genética , Escherichia coli/genética , Plasmídeos/genética , Transfecção
7.
Yeast ; 18(2): 137-50, 2001 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-11169756

RESUMO

A recombinant protein expression system based on Saccharomyces cerevisiae has been used to express malarial vaccine candidate antigens. The antigens so produced have been used in three Phase 1 clinical trials and numerous preclinical non-human primate trials. Further Phase I trials are planned using these candidate vaccine antigens. These molecules were identified as attractive candidates for antimalarial vaccines, as they are all surface-exposed at some stage in the parasite's life cycle. They all share an unusual structural feature: epidermal growth factor (EGF)-like motifs. When these proteins are expressed in our S. cerevisiae expression system, they are produced as a series of stable structural conformers, each with a different disulphide bonding pattern. This leads to both biochemical and, more importantly, antigenic differences between the conformers (e.g. presence or absence of an antibody B cell epitope). These findings have important ramifications for other EGF-domain-containing proteins expressed in S. cerevisiae, or for proteins which contain other cysteine-folding motifs not normally expressed by this organism, both for vaccine production or for research/reagent purposes.


Assuntos
Antígenos de Protozoários/química , Vacinas Antimaláricas/química , Vacinas Sintéticas/química , Animais , Antígenos de Protozoários/imunologia , Antígenos de Protozoários/isolamento & purificação , Ensaios Clínicos Fase I como Assunto , Dissulfetos/química , Fator de Crescimento Epidérmico/química , Humanos , Vacinas Antimaláricas/imunologia , Vacinas Antimaláricas/isolamento & purificação , Plasmodium falciparum/imunologia , Processamento de Proteína Pós-Traducional , Estrutura Secundária de Proteína , Saccharomyces cerevisiae , Vacinas Sintéticas/imunologia , Vacinas Sintéticas/isolamento & purificação
8.
Vaccine ; 19(7-8): 816-24, 2000 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-11115704

RESUMO

Plasmodium falciparum merozoite surface protein 4 (MSP4) is being developed as a component of a subunit vaccine against asexual stages of malaria. Three DNA constructs were produced that induced expression of MSP4 either in the cytoplasm of transfected cells or secreted from cells under the control of the human tissue plasminogen activator (TPA) signal or the native P. falciparum MSP4 signal. Only the construct containing the TPA signal induced detectable antibodies in mice, although gene expression was demonstrated in all constructs and MSP4 was shown to be secreted using either signal by in vitro transient transfection of COS cells. Two recombinant MSP4 proteins that encoded the same sequence as the plasmid DNA were produced in E. coli (EcMSP4-His) and S. cerevisiae (yMSP4-His) and used to raise antibodies in mice. Comparison of the antibodies elicited by these various antigen formulations showed differences in titer, isotype and epitope recognition. The titer of antibodies induced by DNA vaccination was lower than that induced by yMSP4-His, which in turn was lower than that induced by EcMSP4-His. The isotype profiles of the antibodies were also different, the plasmid DNA induced predominantly IgG(2a) responses whereas the two proteins induced predominantly IgG(1) responses. The antibodies induced by DNA and yMSP4-His recognized predominantly the C-terminal epidermal growth factor (EGF)-like domain of the protein, whereas EcMSP4-His induced antibodies recognizing all domains of the protein equally. The antibodies induced by DNA vaccination were directed almost extensively to conformational epitopes so that reactivity with native MSP4 was abolished after disulfide bonds in the protein were disrupted. Antibodies induced by recombinant proteins recognized linear epitopes as well and reactivity to native MSP4 was preserved after reduction and alkylation of parasite proteins.


Assuntos
Antígenos de Protozoários/genética , Antígenos de Protozoários/imunologia , Vacinas Antimaláricas/imunologia , Plasmodium falciparum/genética , Plasmodium falciparum/imunologia , Proteínas de Protozoários/genética , Proteínas de Protozoários/imunologia , Vacinas de DNA/imunologia , Animais , Anticorpos Antiprotozoários/biossíntese , Células COS , Epitopos/genética , Feminino , Expressão Gênica , Genes de Protozoários , Humanos , Vacinas Antimaláricas/genética , Camundongos , Camundongos Endogâmicos C3H , Transfecção , Vacinas de DNA/genética , Vacinas de Subunidades Antigênicas/genética , Vacinas de Subunidades Antigênicas/imunologia
9.
Infect Immun ; 68(12): 6618-23, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11083773

RESUMO

Transmission-blocking vaccines are one strategy for controlling malaria, whereby sexual-stage parasites are inhibited from infecting mosquitoes by human antibodies. To evaluate whether the recently cloned Plasmodium vivax proteins Pvs25 and Pvs28 are candidates for a transmission-blocking vaccine, the molecules were expressed in yeast as secreted recombinant proteins. Mice vaccinated with these proteins adsorbed to aluminum hydroxide developed strong antibody responses against the immunogens, although for Pvs28, this response was genetically restricted. Antisera against both recombinant Pvs25 and Pvs28 recognized the corresponding molecules expressed by cultured sexual-stage parasites isolated from patients with P. vivax malaria. The development of malaria parasites in mosquitoes was completely inhibited when these antisera were ingested with the infected blood meal. Pvs25 and Pvs28, expressed in Saccharomyces cerevisiae, are as yet the only fully characterized transmission-blocking vaccine candidates against P. vivax that induce such a potent antiparasite response.


Assuntos
Anticorpos Antiprotozoários/uso terapêutico , Antígenos de Protozoários/imunologia , Antígenos de Superfície/imunologia , Culicidae/parasitologia , Vacinas Antimaláricas/imunologia , Plasmodium vivax/imunologia , Sequência de Aminoácidos , Animais , Antígenos de Protozoários/genética , Antígenos de Superfície/genética , Clonagem Molecular , Soros Imunes/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Pan troglodytes , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/isolamento & purificação
10.
Infect Immun ; 68(10): 5530-8, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10992450

RESUMO

Each of the four epidermal growth factor (EGF)-like domains of the Plasmodium falciparum sexual-stage antigen Pfs25 has been individually expressed as a yeast-secreted recombinant protein (yEGF1 through yEGF4). All four are recognized by the immune sera of animals and humans vaccinated with TBV25H (the corresponding yeast-secreted full-length recombinant form of Pfs25), with antibody titers to yEGF1 and yEGF2 weakly correlating with the ability of the sera to block the transmission of parasites to the mosquito host. All four proteins are poorly immunogenic in mice vaccinated with aluminum hydroxide-absorbed formulations. However, all four successfully primed the mice to mount an effective secondary antibody response after a single boost with TBV25H. Sera from mice vaccinated with yEGF2-TBV25H completely block the development of oocysts in mosquito midguts in membrane-feeding assays. Further, of the four proteins, only the depletion of antibodies to yEGF2 from the sera of rabbits vaccinated with TBV25H consistently abolished the ability of those sera to block oocyst development. Thus, antibodies to the second EGF-like domain of Pfs25 appear to mediate a very potent blocking activity, even at low titers. Vaccination strategies that target antibody response towards this domain may improve the efficacy of future transmission-blocking vaccines.


Assuntos
Anticorpos Antiprotozoários/imunologia , Vacinas Antimaláricas/imunologia , Malária Falciparum/transmissão , Plasmodium falciparum/imunologia , Proteínas de Protozoários/imunologia , Animais , Anticorpos Antiprotozoários/sangue , Culicidae/parasitologia , Fator de Crescimento Epidérmico/química , Fator de Crescimento Epidérmico/genética , Fator de Crescimento Epidérmico/imunologia , Humanos , Malária Falciparum/parasitologia , Malária Falciparum/prevenção & controle , Camundongos , Plasmodium falciparum/química , Plasmodium falciparum/genética , Plasmodium falciparum/crescimento & desenvolvimento , Estrutura Terciária de Proteína , Proteínas de Protozoários/química , Coelhos , Proteínas Recombinantes/imunologia , Vacinação , Vacinas Sintéticas/imunologia
11.
Mol Microbiol ; 36(1): 1-9, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10760158

RESUMO

Apicomplexan protozoa possess a family of micronemal and cell surface-associated proteins, each comprised a combination of cell-adhesive vertebrate von Willebrand factor (vWF)-like A domains and thrombospondin (TSP) type 1-like domains. The human malaria parasite Plasmodium falciparum has in the extracellular portion of the CS protein TRAP-related protein (CTRP) six tandemly arrayed A domains followed by seven TSP type 1-like domains, whereas a second member of this family, thrombospondin-related anonymous protein (TRAP), contains a single vWF-like A domain and a single TSP type 1-like domain. Here we show that CTRP transcripts are present within the infected mosquito midgut and that CTRP protein is expressed with a punctate distribution and a predominance at the apical end of mosquito midgut-stage ookinetes. This expression pattern is analogous to micronemal expression of TRAP in Plasmodium sporozoites. Disruption of the CTRP gene by homologous recombination in cultures of the human malaria parasite P. falciparum demonstrates that CTRP is essential for mosquito midgut development. Oocyst formation was never observed following membrane feeds of CTRP disruptant lines to Anopheline mosquitoes, despite the development of mature ookinetes. We propose that CTRP is involved in essential recognition or motility processes at the ookinete cell surface within the mosquito midgut.


Assuntos
Anopheles/parasitologia , Genes de Protozoários , Plasmodium falciparum/crescimento & desenvolvimento , Plasmodium falciparum/genética , Proteínas de Protozoários , Receptores de Superfície Celular/genética , Animais , Sistema Digestório/parasitologia , Imunofluorescência , Mutagênese , Fenótipo , Receptores de Superfície Celular/isolamento & purificação
12.
Infect Immun ; 68(5): 3019-22, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10769007

RESUMO

Merozoite surface protein 1 (MSP-1(19)) is a leading malaria vaccine candidate. Specific antibodies contribute to immunity; binding to macrophages is believed to represent the main action of malaria antibodies. We show that an MSP-1(19)-specific immunoglobulin G3 (IgG3) monoclonal antibody can passively transfer protection to mice deficient in the alpha chain of Fc-gammaRI whose macrophages cannot bind IgG3.


Assuntos
Anticorpos Antiprotozoários/imunologia , Imunoglobulina G/imunologia , Malária/prevenção & controle , Proteína 1 de Superfície de Merozoito/imunologia , Plasmodium yoelii/imunologia , Receptores de IgG/imunologia , Animais , Anticorpos Monoclonais/imunologia , Especificidade de Anticorpos , Imunização Passiva/métodos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fragmentos de Peptídeos/imunologia , Receptores de IgG/genética
13.
J Biol Chem ; 275(14): 10331-41, 2000 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-10744721

RESUMO

The Plasmodium ookinete produces chitinolytic activity that allows the parasite to penetrate the chitin-containing peritrophic matrix surrounding the blood meal in the mosquito midgut. Since the peritrophic matrix is a physical barrier that the parasite must cross to invade the mosquito, and the presence of allosamidin, a chitinase inhibitor, in a blood meal prevents the parasite from invading the midgut epithelium, chitinases (3.2.1.14) are potential targets of malaria parasite transmission-blocking interventions. We have purified a chitinase of the avian malaria parasite Plasmodium gallinaceum and cloned the gene, PgCHT1, encoding it. PgCHT1 encodes catalytic and substrate-binding sites characteristic of family 18 glycohydrolases. Expressed in Escherichia coli strain AD494 (DE3), recombinant PgCHT1 was found to hydrolyze polymeric chitin, native chitin oligosaccharides, and 4-methylumbelliferone derivatives of chitin oligosaccharides. Allosamidin inhibited recombinant PgCHT1 with an IC(50) of 7 microM and differentially inhibited two chromatographically separable P. gallinaceum ookinete-produced chitinase activities with IC(50) values of 7 and 12 microM, respectively. These two chitinase activities also had different pH activity profiles. These data suggest that the P. gallinaceum ookinete uses products of more than one chitinase gene to initiate mosquito midgut invasion.


Assuntos
Quitinases/genética , Quitinases/metabolismo , Culicidae/parasitologia , Plasmodium gallinaceum/fisiologia , Sequência de Aminoácidos , Animais , Galinhas , Quitinases/isolamento & purificação , Sequência Consenso , Sistema Digestório/parasitologia , Células Epiteliais/enzimologia , Regulação da Expressão Gênica no Desenvolvimento , Regulação Enzimológica da Expressão Gênica , Humanos , Cinética , Malária Aviária , Dados de Sequência Molecular , Plasmodium gallinaceum/genética , Plasmodium gallinaceum/patogenicidade , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos
14.
Infect Immun ; 68(4): 2215-23, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10722622

RESUMO

The immunogenicity and protective efficacy of four versions of recombinant C-terminal 19-kDa epidermal growth factor-like region of the major surface protein 1 (rMSP1(19)) of Plasmodium falciparum was studied in Aotus monkeys. Vaccination with each of the four rMSP1(19) constructs elicited high levels of antibodies to MSP1(19) but only one construct, the 19-kDa fragment expressed as a secreted fusion protein from Saccharomyces cerevisiae (yP30P2MSP1(19)), induced a high degree of protective immunity in Aotus nancymai against lethal P. falciparum challenge. Protective formulation required Freund's adjuvant; vaccination with yP30P2MSP1(19) in six other adjuvants that are suitable for human use induced lower levels of antibody response and no protection. These results emphasize the need to continue the search for an adjuvant that is comparable to Freund's adjuvant in potency and is safe for use in humans.


Assuntos
Aotidae/parasitologia , Proteína 1 de Superfície de Merozoito/imunologia , Plasmodium falciparum/imunologia , Vacinas Sintéticas , Adjuvantes Imunológicos , Animais , Sistemas de Liberação de Medicamentos , Ensaio de Imunoadsorção Enzimática , Epitopos , Feminino , Técnica Indireta de Fluorescência para Anticorpo , Adjuvante de Freund/imunologia , Malária Falciparum/prevenção & controle , Masculino , Proteínas Recombinantes de Fusão/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Toxoide Tetânico/imunologia , Fatores de Tempo
15.
Infect Immun ; 68(3): 1418-27, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10678955

RESUMO

Protection against a lethal challenge infection of Plasmodium falciparum was elicited in malaria-naive Aotus vociferans monkeys by vaccination with the C terminus 19-kDa protein of the major merozoite surface protein (MSP-1(19)) fused to tetanus toxoid universal T-cell epitopes P30 and P2. Three of four monkeys were protected against a 10(4)-parasite challenge. Four monkeys were challenged with 10(5) parasites; one self-cured the infection, two were protected against high parasitemia (<2%) but were treated for severe anemia (hematocrit of <25%), and the fourth was not protected. In this model system, anemia appears to be a manifestation of incomplete protection (prolonged low-level parasitemia). Enzyme-linked immunosorbent assay (ELISA) antibody titers correlated with protection. Antibodies from some protected monkeys inhibited secondary processing of MSP-1(42) to MSP-1(33) and MSP-1(19). To mimic the repeated reinfections seen in regions where malaria is endemic, a second malaria parasite challenge was administered 4 months later. All P30P2MSP-1(19)-vaccinated monkeys were protected; thus, a single challenge infection may underestimate vaccine efficacy. ELISA antibody titers correlated with protection against a second infection but had decreased compared to the first challenge. As most target populations for asexual blood-stage malaria vaccines will have been exposed to malaria parasites, a malaria parasite-exposed monkey was vaccinated with P30P2MSP-1(19). This monkey was completely protected, while a malaria parasite-naive P30P2MSP-1(19)-vaccinated monkey self-cured a low-grade parasitemia. Prior malaria parasite infection primed the production of anti-native MSP-1(19) antibodies, which were boosted by vaccination with recombinant P30P2MSP-1(19). Preliminary data suggest that immunogenicity studies of vaccines designed for malaria parasite-exposed populations should also be conducted in malaria parasite-exposed subjects.


Assuntos
Vacinas Antimaláricas/imunologia , Proteína 1 de Superfície de Merozoito/imunologia , Plasmodium falciparum/imunologia , Vacinas Sintéticas/imunologia , Animais , Anticorpos Antiprotozoários/sangue , Aotus trivirgatus , Ensaio de Imunoadsorção Enzimática , Feminino , Malária Falciparum/prevenção & controle , Masculino , Vacinação
16.
Mol Microbiol ; 35(1): 6-14, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10632873

RESUMO

Malaria infects over 200 million individuals and kills 2 million young children every year. Understanding the biology of malarial parasites will be facilitated by DNA microarray technology, which can track global changes in gene expression under different physiological conditions. However, genomes of Plasmodium sp. (and many other important pathogenic organisms) remain to be fully sequenced so, currently, it is not possible to construct gene-specific microarrays representing complete malarial genomes. In this study, 3648 random inserts from a Plasmodium falciparum mung bean nuclease genomic library were used to construct a shotgun DNA microarray. Through differential hybridization and sequencing of relevant clones, large differences in gene expression were identified between the blood stage trophozoite form of the malarial parasite and the sexual stage gametocyte form. The present study lengthens our list of stage-specific transcripts in malaria by at least an order of magnitude above all previous studies combined. The results offer an unprecedented number of leads for developing transmission blocking agents and for developing vaccines directed at blood stage antigens. A significant fraction of the stage-selective transcripts had no sequence homologues in the current genome data bases, thereby underscoring the importance of the shotgun approach. The malarial shotgun microarray will be useful for unravelling additional important aspects of malaria biology and the general approach may be applied to any organism, regardless of how much of its genome is sequenced.


Assuntos
DNA de Protozoário/genética , Regulação da Expressão Gênica no Desenvolvimento , Plasmodium falciparum/genética , Animais , Sequência de Bases , Primers do DNA , Genoma de Protozoário , Hibridização de Ácido Nucleico , Reação em Cadeia da Polimerase , Transcrição Gênica
17.
Am J Trop Med Hyg ; 62(6): 720-5, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11304063

RESUMO

Anti-Pv200 antibody levels were assessed in samples from endemic areas of Plasmodium vivax malaria in the Republic of Korea (ROK), using an indirect enzyme-linked immunosorbent assay (ELISA) method. Asymptomatic carriers of P. vivax were detected using nested polymerase chain reaction (PCR) of blood samples. Anti-Pv200 antibody levels in 20 vivax malaria patients (optical density +/- standard deviation [OD +/- SD] values 1.85 +/- 0.29 of IgG isotype and 1.33 +/- 1.33 of IgM isotype) were markedly higher than those of uninfected, malaria-naive controls (0.08 +/- 0.16 of IgG isotype and 0.04 +/- 0.04 of IgM isotype). Antibody levels for 7 out of 8 soldiers with a recent malaria infection were sustained above the cut-off values for 4 months after successful treatment. Analysis of serum collected from 40 healthy, asymptomatic soldiers who had a P. vivax malaria attack within 3 months after our sampling, revealed 11 antibody-positive samples (27.5%), compared to 5 positive samples (12.5%) collected from a random selection of 40 soldiers. Among a larger pool of 1,713 soldiers who had served in high-risk areas for P. vivax transmission, 15% were antibody positive. Among 1,000 blood samples from asymptomatic soldiers who had served in the high-risk areas, 4 samples (0.4%) were parasite positive, as determined by nested PCR. Our results show that anti-Pv200 antibody levels can provide useful information in the late diagnosis of P. vivax malaria infection in a previously naive population and also in large seroepidemiologic studies. Furthermore, our results suggest that asymptomatic P. vivax carriers could be important in the current outbreak of malaria in Korea.


Assuntos
Anticorpos Antiprotozoários/sangue , Surtos de Doenças , Malária Vivax/epidemiologia , Malária Vivax/imunologia , Militares , Plasmodium vivax/imunologia , Animais , Antígenos de Protozoários/imunologia , Antígenos de Superfície/imunologia , Portador Sadio/parasitologia , DNA de Protozoário/análise , Doenças Endêmicas , Humanos , Coreia (Geográfico)/epidemiologia , Malária Vivax/parasitologia , Plasmodium vivax/genética , Plasmodium vivax/isolamento & purificação , Reação em Cadeia da Polimerase/métodos , Proteínas de Protozoários/imunologia , Estudos Soroepidemiológicos
18.
Am J Trop Med Hyg ; 60(3): 350-6, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10466960

RESUMO

Saimiri boliviensis monkeys were immunized with the yeast-expressed recombinant protein yP2P30Pv200(19). The antigen consisted of the C-terminus (amino acid Asn1622-Ser1729) of the merozoite surface protein 1 of the Plasmodium vivax Salvador I strain. Two universal T helper cell epitopes (P2 and P30) of tetanus toxin and six histidine residues for purification purposes were attached to the N- and C-termini, respectively. Four groups of five monkeys were given three immunizations at four-week intervals with either 250 microg of yP2P30Pv200(19) formulated with nonionic block copolymer P1005, 250 microg of antigen adsorbed to alum, 250 microg of antigen in phosphate-buffered saline (PBS), or PBS alone. Five weeks after the last immunization, each animal was inoculated with 100,000 parasitized erythrocytes of the Salvador I strain of P. vivax. Animals were splenectomized one week after challenge to increase parasite densities; after seven weeks of infection, animals were treated. Eighteen weeks later, the animals were rechallenged with the homologous parasite. Following the first challenge, three monkeys immunized with the antigen with P1005 were protected; no animals were protected from rechallenge. One monkey immunized with yP2P30Pv200(19) with alum was protected; no protection was seen after rechallenge. Two monkeys immunized with antigen alone were protected; none were protected from rechallenge. One control animal had a low parasite count following primary infection; none were protected against rechallenge. Adverse reactions were only observed with animals receiving P1005. It is proposed that splenectomy of the monkeys prevented adequate assessment of the efficacy of this antigen. Identification of a monkey host that supports high density parasitemia without splenectomy appears needed before further testing of blood-stage vaccines against P. vivax.


Assuntos
Vacinas Antimaláricas/normas , Malária Vivax/prevenção & controle , Proteína 1 de Superfície de Merozoito/imunologia , Plasmodium vivax/imunologia , Vacinas Sintéticas/normas , Adjuvantes Imunológicos/uso terapêutico , Animais , Antimaláricos/uso terapêutico , Modelos Animais de Doenças , Feminino , Imunização , Vacinas Antimaláricas/imunologia , Malária Vivax/imunologia , Masculino , Mefloquina/uso terapêutico , Parasitemia , Saimiri , Esplenectomia , Vacinas Sintéticas/imunologia
20.
J Immunol ; 162(12): 7309-14, 1999 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-10358180

RESUMO

Vaccination of mice with the leading malaria vaccine candidate homologue, the 19-kDa carboxyl terminus of merozoite surface protein-1 (MSP119), results in sterile immunity to Plasmodium yoelii, with no parasites detected in blood. Although such immunity depends upon high titer Abs at challenge, high doses of immune sera transferred into naive mice reduce parasitemia (and protect from death) but do not result in a similar degree of protection (with most mice experiencing high peak parasitemias); this finding suggests that ongoing parasite-specific immune responses postchallenge are essential. We analyzed this postchallenge response by transferring Abs into manipulated but malaria-naive mice and observed that Abs cannot protect SCID, nude, CD4+ T cell-depleted, or B cell knockout mice, with all mice dying. Thus, in addition to the Abs that develop following MSP119 vaccination, a continuing active immune response postchallenge is required for protection. MSP119-specific Abs can adoptively transfer protection to strains of mice that are not protected following vaccination with MSP119, suggesting that the Ags targeted by the immune response postchallenge include Ags apart from MSP119. These data have important implications for the development of a human malaria vaccine.


Assuntos
Anticorpos Antiprotozoários/administração & dosagem , Linfócitos B/imunologia , Malária/imunologia , Proteína 1 de Superfície de Merozoito/imunologia , Fragmentos de Peptídeos/imunologia , Plasmodium yoelii/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Anticorpos Antiprotozoários/uso terapêutico , Especificidade de Anticorpos , Linfócitos B/metabolismo , Linfócitos B/parasitologia , Imunidade Ativa , Imunização Passiva , Injeções Intraperitoneais , Malária/parasitologia , Malária/prevenção & controle , Vacinas Antimaláricas/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Nus , Camundongos SCID , Peso Molecular , Linfócitos T Auxiliares-Indutores/metabolismo , Linfócitos T Auxiliares-Indutores/parasitologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA