Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros










Intervalo de año de publicación
1.
J Exp Biol ; 206(Pt 21): 3789-802, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14506214

RESUMEN

Recent advances in the fields of genomics, proteomics and molecular immunology offer tremendous opportunities for the development of novel interventions against public health threats, including malaria. However, there is currently no algorithm that can effectively identify the targets of protective T cell or antibody responses from genomic data. Furthermore, the identification of antigens that will stimulate the most effective immunity against the target pathogen is problematic, particularly if the genome is large. Malaria is an attractive model for the development and validation of approaches to translate genomic information to vaccine development because of the critical need for effective anti-malarial interventions and because the Plasmodium parasite is a complex multistage pathogen targeted by multiple immune responses. Sterile protective immunity can be achieved by immunization with radiation-attenuated sporozoites, and anti-disease immunity can be induced in residents in malaria-endemic areas. However, the 23 Mb Plasmodium falciparum genome encodes more than 5,300 proteins, each of which is a potential target of protective immune responses. The current generation of subunit vaccines is based on a single or few antigens and therefore might elicit too narrow a breadth of response. We are working towards the development of a new generation vaccine based on the presumption that duplicating the protection induced by the whole organism may require a vaccine nearly as complex as the organism itself. Here, we present our strategy to exploit the genomic sequence of P. falciparum for malaria vaccine development.


Asunto(s)
Antígenos de Protozoos/genética , Expresión Génica , Genoma de Protozoos , Vacunas contra la Malaria/genética , Modelos Inmunológicos , Plasmodium falciparum/genética , Animales , Epítopos/genética , Sistemas de Lectura Abierta/genética , Reacción en Cadena de la Polimerasa , Análisis por Matrices de Proteínas , Vacunas Sintéticas
2.
Mol Biochem Parasitol ; 118(2): 233-45, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11738713

RESUMEN

The detection and quantitation of blood stage parasitaemia is typically used as a surrogate endpoint for estimating the efficacy of vaccines targeted against the hepatic stage, as well as the erythrocytic stage, of the parasite. However, this does not provide an adequate means of evaluating the efficacy of vaccines, which may be only partially effective at the liver-stage. This is a particular concern for effective evaluation of immune enhancement strategies for candidate pre-erythrocytic stage vaccines. Here, we have developed and validated a method for detecting and quantitating liver stage parasites, using the TaqMan fluorescent real-time quantitative PCR system (PE Applied Biosystems). This method uses TaqMan primers designed to the Plasmodium yoelii 18S rRNA gene and rodent GAPDH to amplify products from infected mouse liver cDNA. The technique is highly reproducible as demonstrated with plasmid controls and capable of efficiently quantitating liver-stage parasite burden following a range of sporozoite challenge doses in strains of mice, which differ in their susceptibility to sporozoite infection. We have further demonstrated the capacity of this technique to evaluate the efficacy of a range of pre-erythrocytic stage vaccines. Our data establish this quantitative real-time PCR assay to be a fast and reproducible way of accurately assessing liver stage parasite burden and vaccine efficacy in rodent malaria models.


Asunto(s)
Hígado/parasitología , Vacunas contra la Malaria , Malaria/prevención & control , Plasmodium yoelii/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Animales , Anticuerpos Monoclonales/inmunología , ADN Ribosómico/análisis , Eritrocitos/parasitología , Femenino , Genes de ARNr , Inmunización Pasiva , Malaria/parasitología , Vacunas contra la Malaria/administración & dosificación , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Plasmodium yoelii/genética , Plasmodium yoelii/crecimiento & desarrollo , Plasmodium yoelii/patogenicidad , ARN Ribosómico 18S/genética , Análisis de Secuencia de ADN , Polimerasa Taq/metabolismo , Vacunación , Vacunas de ADN/administración & dosificación
3.
Immunogenetics ; 53(4): 315-28, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11491535

RESUMEN

The finding that a single administration of select recombinant human cytokines to nonhuman primates leads to potent cytokine-neutralizing antibody responses in the heterologous host despite >95% homology at the nucleotide and protein level prompted our laboratory to clone, sequence, and prepare recombinant nonhuman primate cytokines, chemokines, growth factors, and other immunoregulatory molecules. In the present report, we present findings on the gene sequences encoding the nonhuman primate homologues of human CD80, CD86, their ligands CD28 and CD152, CD154, CD95, and CD95-L from rhesus macaques and for phylogenetic analysis from pig-tailed macaques, African sooty mangabey monkeys, baboons, and vervets as well as select molecules from the New World aotus and marmoset monkeys. With the exception of CD95, the homology between nonhuman primate and human co-stimulatory molecules was above 95%. In contrast, CD95 was only 89.2% homologous to human CD95, but the differences were essentially found in the transmembrane and intracellular (death) domains. The extracellular portion of CD95 was more homologous which was in accordance with approximately 98% homology between Old World monkey and human CD95-L. In general, sequences from the New World monkey species appeared equidistant to sequences from Old World species and humans in terms of homology suggesting distinct evolutionary patterns. Of interest was the isolation of various splice variants of monkey CD86, CD152 (CTLA-4), CD154, and CD95 transcripts. This is also the first report documenting the occurrence of natural CD86 variants with deleted transmembrane domains, found both in sooty mangabeys and baboon RNA samples. Monkey CD95 showed various deletions and addition of residues in the transmembrane and intracytoplasmic domains compared with human CD95 and between Old and New World species. Subcloning of rhesus CD154 into an expression vector demonstrated expression of a functional protein in cell culture. The other genes are being cloned into expression vectors for the preparation and biological characterization of the nonhuman primate molecules. These investigations will provide novel reagents for in vivo use as immunomodulatory reagents in nonhuman primates in studies which may provide a rationale for their use in humans.


Asunto(s)
Antígenos CD/genética , Antígeno B7-1/genética , Haplorrinos/genética , Inmunoconjugados , Glicoproteínas de Membrana/genética , Receptor fas/genética , Abatacept , Secuencia de Aminoácidos , Animales , Antígenos de Diferenciación/genética , Antígeno CTLA-4 , Clonación Molecular , Evolución Molecular , Proteína Ligando Fas , Leucocitos Mononucleares/citología , Datos de Secuencia Molecular , Análisis de Secuencia de ADN , Homología de Secuencia de Aminoácido
4.
J Immunol ; 165(7): 3772-81, 2000 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-11034382

RESUMEN

Plasmid-encoded GM-CSF (pGM-CSF) is an adjuvant for genetic vaccines; however, little is known about how pGM-CSF enhances immunogenicity. We now report that pGM-CSF injected into mouse muscle leads to a local infiltration of potential APCs. Infiltrates reached maximal size on days 3 to 5 after injection and appeared in several large discrete clusters within the muscle. Immunohistological studies in muscle sections from mice injected with pGM-CSF showed staining of cells with the macrophage markers CD11b, Mac-3, IA(d)/E(d) and to the granulocyte marker GR-1 from day 1 through day 14. Cells staining with the dendritic cell marker CD11c were detected only on days 3 to 5. Muscles injected with control plasmids did not stain for CD11c but did stain for CD11b, Mac-3, IA(d)/E(d), and GR-1. No staining was observed with the APC activation markers, B7.1 or CD40, or with markers for T or B cells. These findings are consistent with the infiltrating cells in the pGM-CSF-injected muscles being a mixture of neutrophils, macrophages, and immature dendritic cells and suggest that the i.m. APCs may be enhancing immune responses to coinjected plasmid Ags. This hypothesis is supported by data showing that 1) separation of injections with pGM-CSF and Ag-expressing plasmid into different sites did not enhance immune responses and 2) immune enhancement was associated with the presence of CD11c+ cells in the infiltrates. Thus, pGM-CSF enhancement may depend on APC recruitment to the i.m. site of injection.


Asunto(s)
Movimiento Celular/inmunología , Células Dendríticas/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/administración & dosificación , Factor Estimulante de Colonias de Granulocitos y Macrófagos/biosíntesis , Músculo Esquelético/inmunología , Plásmidos/administración & dosificación , Plásmidos/inmunología , Vacunas de ADN/administración & dosificación , Vacunas de ADN/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/genética , Adyuvantes Inmunológicos/metabolismo , Animales , Antígenos de Protozoos/administración & dosificación , Antígenos de Protozoos/genética , Antígenos de Protozoos/inmunología , Diferenciación Celular/inmunología , Células Dendríticas/química , Células Dendríticas/metabolismo , Células Dendríticas/patología , Relación Dosis-Respuesta Inmunológica , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Inmunidad Celular/genética , Inmunohistoquímica , Inyecciones Intramusculares , Inyecciones Intraventriculares , Integrina alfaXbeta2/biosíntesis , Cinética , Ganglios Linfáticos/química , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Ratones , Ratones Endogámicos BALB C , Músculo Esquelético/citología , Plásmidos/metabolismo , Plasmodium yoelii/genética , Plasmodium yoelii/inmunología , Vacunas de ADN/metabolismo
5.
J Gen Virol ; 81(Pt 7): 1659-67, 2000 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10859370

RESUMEN

A candidate DNA vaccine expressing dengue virus type 1 pre-membrane and envelope proteins was used to immunize rhesus macaques. Monkeys were immunized intramuscularly (i.m.) or intradermally (i.d.) by three or four 1 mg doses of vaccine, respectively. Monkeys that were inoculated i.m. seroconverted more quickly and had higher antibody levels than those that were inoculated i.d. The sera exhibited virus-neutralizing activity, which declined over time. Four of the eight i.m.-inoculated monkeys were protected completely from developing viraemia when challenged 4 months after the last dose with homologous dengue virus. The other four monkeys had reduced viraemia compared with the control immunized monkeys. The i.d. -inoculated monkeys showed no reduction in viraemia when challenged with the virus. All vaccinated monkeys showed an anamnestic antibody response, indicating that they had established immunological memory. Vaccine-induced antibody had an avidity index similar to that of antibody induced by virus infection; however, no clear correlation was apparent between antibody avidity and virus neutralization titres.


Asunto(s)
Virus del Dengue/inmunología , Vacunas de ADN/inmunología , Vacunas Virales/inmunología , Animales , Afinidad de Anticuerpos , Inmunización , Memoria Inmunológica , Activación de Linfocitos , Macaca mulatta , Linfocitos T/inmunología
6.
Vaccine ; 18(7-8): 703-10, 1999 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-10547430

RESUMEN

A plasmid DNA vaccine encoding the circumsporozoite protein of malaria (pCSP) induces protective immunity in adult mice but persistent tolerance when administered to neonates. In an effort to improve antigen presenting cell (APC) function in newborns, we co-administered pCSP with a plasmid encoding granulocyte-macrophage colony stimulating factor (pGMCSF). This combination of plasmids prevented the development of neonatal tolerance, instead eliciting a primary IgG anti-CSP immune response. Mice primed as neonates and boosted as adults mounted anamnestic responses characterized by high serum antibody titers, cytotoxic T-cell activity and antigen-specific interferon gamma (IFNgamma) production. Neonatal administration of pGMCSF accelerated the maturation of local dendritic cells, suggesting that APC function plays a key role in determining whether tolerance or immunity results from neonatal exposure to antigen.


Asunto(s)
Animales Recién Nacidos/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Tolerancia Inmunológica , Vacunas de ADN/inmunología , Animales , Células Presentadoras de Antígenos/citología , Células Presentadoras de Antígenos/inmunología , Antígenos de Protozoos/genética , Antígenos de Protozoos/inmunología , Diferenciación Celular/inmunología , Células Dendríticas/citología , Células Dendríticas/inmunología , Femenino , Humanos , Tolerancia Inmunológica/genética , Interferón gamma/biosíntesis , Interferón gamma/inmunología , Ratones , Ratones Endogámicos BALB C , Plásmidos/administración & dosificación , Plásmidos/inmunología , Plasmodium yoelii/genética , Plasmodium yoelii/inmunología , Proteínas Protozoarias/genética , Proteínas Protozoarias/inmunología , Linfocitos T Citotóxicos/inmunología , Vacunas de ADN/genética
7.
Gene Ther ; 6(2): 237-44, 1999 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10435108

RESUMEN

Cytokine-encoding DNA plasmids can act as 'genetic adjuvants', improving the immune response stimulated by co-administered DNA vaccines. We examined whether plasmids encoding the Th1 cytokine IFN gamma (pIFN gamma) or the Th2 cytokine IL-4 (pIL-4) have long-term effects on immune homeostasis when administered to adult mice, or alter immune maturation in neonates. Both plasmids boosted immunity against a co-administered vaccine, with pIFN gamma promoting the development of a Th1 response (characterized by the production of IgG2a antibodies), and pIL-4 preferentially stimulating a Th2 response (characterized by increased IgG1 antibody production). Both pIFN gamma and pIL-4 influenced the ratio of cells actively secreting Th1 versus Th2 cytokines, consistent with an effect on Th cell maturation. Interestingly, this effect persisted for only a few weeks and was not magnified by repeated plasmid administration. Cytokine-encoding plasmids had no long-term effect on the immune response of newborn or adult mice to subsequent antigenic stimulation, nor did they selectively induce the production of pathogenic anti-DNA autoantibodies. These results suggest cytokine-encoding plasmids may be safe as immune adjuvants.


Asunto(s)
Adyuvantes Inmunológicos , Terapia Genética/métodos , Interferón gamma/genética , Interleucina-4/genética , Plásmidos/administración & dosificación , Vacunas de ADN/administración & dosificación , Animales , Animales Recién Nacidos , Anticuerpos/sangre , Ensayo de Inmunoadsorción Enzimática/métodos , Inmunoglobulina G , Ratones , Ratones Endogámicos BALB C , Células TH1/inmunología , Células Th2/inmunología
8.
J Immunol ; 162(7): 3814-8, 1999 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-10201898

RESUMEN

A plasmid DNA vaccine encoding the circumsporozoite protein of malaria (pCSP) induces tolerance rather than immunity when administered to newborn mice. We find that this tolerance persists for >1 yr after neonatal pCSP administration and interferes with the induction of protective immunity in animals challenged with live sporozoites. Susceptibility to tolerance induction wanes rapidly with age, disappearing within 1 wk of birth. Higher doses of plasmid are more tolerogenic, and susceptibility to tolerance is not MHC-restricted. CD8+ T cells from tolerant mice suppress the in vitro Ag-specific immune response of cells from adult mice immunized with pCSP. Similarly, CD8+ T cells from tolerant mice transfer nonresponsiveness to naive syngeneic recipients. These findings clarify the cellular basis and factors contributing to the development of DNA vaccine-induced neonatal tolerance.


Asunto(s)
Animales Recién Nacidos/inmunología , Tolerancia Inmunológica , Plásmidos/inmunología , Vacunas de ADN/inmunología , Animales , Animales Recién Nacidos/genética , Susceptibilidad a Enfermedades , Relación Dosis-Respuesta Inmunológica , Femenino , Tolerancia Inmunológica/genética , Inmunidad Celular/genética , Inyecciones Intramusculares , Malaria/genética , Malaria/inmunología , Ratones , Ratones Endogámicos A , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Plásmidos/administración & dosificación , Plásmidos/genética , Plasmodium yoelii/genética , Plasmodium yoelii/inmunología , Proteínas Protozoarias/genética , Proteínas Protozoarias/inmunología , Vacunas de ADN/administración & dosificación , Vacunas de ADN/genética
9.
Science ; 282(5388): 476-80, 1998 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-9774275

RESUMEN

CD8+ cytotoxic T lymphocytes (CTLs) are critical for protection against intracellular pathogens but often have been difficult to induce by subunit vaccines in animals. DNA vaccines elicit protective CD8+ T cell responses. Malaria-naïve volunteers who were vaccinated with plasmid DNA encoding a malaria protein developed antigen-specific, genetically restricted, CD8+ T cell-dependent CTLs. Responses were directed against all 10 peptides tested and were restricted by six human lymphocyte antigen (HLA) class I alleles. This first demonstration in healthy naïve humans of the induction of CD8+ CTLs by DNA vaccines, including CTLs that were restricted by multiple HLA alleles in the same individual, provides a foundation for further human testing of this potentially revolutionary vaccine technology.


Asunto(s)
Vacunas contra la Malaria/inmunología , Plasmodium falciparum/inmunología , Proteínas Protozoarias/genética , Proteínas Protozoarias/inmunología , Linfocitos T Citotóxicos/inmunología , Vacunas de ADN/inmunología , Adulto , Animales , Antígenos de Protozoos/genética , Antígenos de Protozoos/inmunología , Femenino , Genes MHC Clase I , Antígenos HLA/genética , Humanos , Esquemas de Inmunización , Vacunas contra la Malaria/genética , Masculino , Plasmodium falciparum/genética , Vacunación
10.
J Immunol ; 161(5): 2325-32, 1998 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-9725227

RESUMEN

Using the murine parasite Plasmodium yoelii (Py) as a model for malaria vaccine development, we have previously shown that a DNA plasmid encoding the Py circumsporozoite protein (PyCSP) can protect mice against sporozoite infection. We now report that mixing a new plasmid PyCSP1012 with a plasmid encoding murine granulocyte-macrophage colony-stimulating factor (GM-CSF) increases protection against malaria, and we have characterized in detail the increased immune responses due to GM-CSF. PyCSP1012 plasmid alone protected 28% of mice, and protection increased to 58% when GM-CSF was added (p < 0.0001). GM-CSF plasmid alone did not protect, and control plasmid expressing inactive GM-CSF did not enhance protection. GM-CSF plasmid increased Abs to PyCSP of IgG1, IgG2a, and IgG2b isotypes, but not IgG3 or IgM. IFN-gamma responses of CD8+ T cells to the PyCSP 280-288 amino acid epitope increased but CTL activity did not change. The most dramatic changes after adding GM-CSF plasmid were increases in Ag-specific IL-2 production and CD4+ T cell proliferation. We hypothesize that GM-CSF may act on dendritic cells to enhance presentation of the PyCSP Ag, with enhanced IL-2 production and CD4+ T cell activation driving the increases in Abs and CD8+ T cell function. Recombinant GM-CSF is already used in humans for medical purposes, and GM-CSF protein or plasmids may be useful as enhancers of DNA vaccines.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Vacunas contra la Malaria/inmunología , Malaria/inmunología , Malaria/prevención & control , Plásmidos/inmunología , Plasmodium yoelii/inmunología , Vacunas de ADN/inmunología , Animales , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Citocinas/metabolismo , Citotoxicidad Inmunológica/efectos de los fármacos , Citotoxicidad Inmunológica/genética , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Inmunoglobulina G/biosíntesis , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/genética , Malaria/genética , Vacunas contra la Malaria/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Plásmidos/farmacología , Plasmodium yoelii/genética , Plasmodium yoelii/crecimiento & desarrollo , Vacunas de ADN/genética
11.
Infect Immun ; 66(9): 4193-202, 1998 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-9712767

RESUMEN

CD8(+) T cells have been implicated as critical effector cells in protective immunity against malaria parasites developing within hepatocytes. A vaccine that protects against malaria by inducing CD8(+) T cells will probably have to include multiple epitopes on the same protein or different proteins, because of parasite polymorphism and genetic restriction of T-cell responses. To determine if CD8(+) T-cell responses against multiple P. falciparum proteins can be induced in primates by immunization with plasmid DNA, rhesus monkeys were immunized intramuscularly with a mixture of DNA plasmids encoding four P. falciparum proteins or with individual plasmids. All six monkeys immunized with PfCSP DNA, seven of nine immunized with PfSSP2 DNA, and five of six immunized with PfExp-1 or PfLSA-1 DNA had detectable antigen-specific cytotoxic T lymphocytes (CTL) after in vitro restimulation of peripheral blood mononuclear cells. CTL activity was genetically restricted and dependent on CD8(+) T cells. By providing the first evidence for primates that immunization with a mixture of DNA plasmids induces CD8(+) T-cell responses against all the components of the mixture, these studies provide the foundation for multigene immunization of humans.


Asunto(s)
ADN Protozoario/inmunología , Vacunas contra la Malaria/inmunología , Malaria Falciparum/inmunología , Plasmodium falciparum/inmunología , Linfocitos T Citotóxicos/inmunología , Vacunas de ADN/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Antiprotozoarios/inmunología , Antígenos de Protozoos/genética , Antígenos de Protozoos/inmunología , Linfocitos T CD8-positivos/inmunología , Humanos , Interferón gamma/genética , Macaca mulatta , Vacunas contra la Malaria/genética , Malaria Falciparum/genética , Malaria Falciparum/prevención & control , Datos de Secuencia Molecular , Plásmidos , Primates , Proteínas Protozoarias/genética , Proteínas Protozoarias/inmunología , ARN Mensajero , Linfocitos T Citotóxicos/parasitología , Vacunación
12.
Eur J Immunol ; 27(8): 1952-7, 1997 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9295031

RESUMEN

In this study, we have investigated the extent of natural polymorphism in the CD8+ cytotoxic T lymphocyte (CTL) determinant (amino acids 368-390) of circumsporozoite (CS) protein of Plasmodium falciparum field isolates from a holoendemic region of Kenya, and determined how this variation affects the CTL reactivities in clinically immune adults and binding specificities to human histocompatibility leukocyte antigen (HLA)-B35. Among the eight variant sequences that were found in this region, four were new and not seen in parasites from other geographical regions. When synthetic peptides corresponding to the eight variants were used to test the presence of CTL response in different donors, a different spectrum of CTL reactivity to these variants was noticed. While CTL from some donors recognized the P1 sequence (the most prevalent type of sequence) but not P8 (another major variant), other donors showed a reverse pattern of reactivity. Although none of the donors was able to recognize all the variants, CTL responses to all the eight variant sequences were found in this population. An octamer peptide with P1 sequence KPKDELDY in this polymorphic determinant was known to bind HLA-B35. When we tested the effect of natural variation in this octamer sequence on HLA-B35 binding, it became evident that SP13 with D --> N substitution retained its binding specificity to HLA-B35. On the other hand, the SP12 octamer sequence which had two substitutions did not bind HLA-B35. The most interesting finding was the observation that a D --> G substitution at position 374 rescued the binding ability of SP14, which otherwise could not bind to this HLA molecule due to E --> Q amino acid substitution at position 372. To our knowledge, this is the first demonstration showing that a natural polymorphism can rescue the binding specificity to an HLA-class I molecule that was lost due to another natural amino acid substitution. Altogether, these results demonstrate that natural polymorphism in the CS protein affects both the CTL reactivity and the ability to bind to HLA-B35.


Asunto(s)
Antígenos de Protozoos/metabolismo , Antígeno HLA-B35/metabolismo , Malaria Falciparum/inmunología , Plasmodium falciparum/inmunología , Proteínas Protozoarias/inmunología , Proteínas Protozoarias/metabolismo , Linfocitos T Citotóxicos/inmunología , Adulto , Secuencia de Aminoácidos , Animales , Antígenos de Protozoos/genética , Secuencia de Bases , Linfocitos T CD8-positivos/inmunología , ADN Protozoario/genética , Epítopos/genética , Epítopos/metabolismo , Variación Genética , Humanos , Kenia , Malaria Falciparum/parasitología , Datos de Secuencia Molecular , Plasmodium falciparum/genética , Unión Proteica , Proteínas Protozoarias/genética
13.
Immunol Cell Biol ; 75(4): 376-81, 1997 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9315481

RESUMEN

In mid 1997 the first malaria DNA vaccine will enter clinical trials. This single gene DNA vaccine encoding the Plasmodium falciparum circumsporozoite protein (PfCSP) will be studied for safety and immunogenicity. If these criteria are met, a multi-gene DNA vaccine designed to induce protective CD8+ T cell responses against P. falciparum infected hepatocytes will be subsequently assessed for safety, immunogenicity and capacity to protect immunized volunteers against experimental challenge with P. falciparum sporozoites. Our perspectives on malaria vaccine development in general, and on a multi-gene DNA vaccine in particular, have been recently reviewed. Herein, we review the rationale and experimental foundation for the anticipated P. falciparum DNA vaccine trials.


Asunto(s)
Antígenos de Protozoos/inmunología , Malaria/prevención & control , Vacunación/métodos , Vacunas de ADN/uso terapéutico , Animales , Linfocitos T CD8-positivos/inmunología , Ensayos Clínicos como Asunto , Inmunidad Activa , Hepatopatías/inmunología , Hepatopatías/microbiología , Hepatopatías/prevención & control , Macaca mulatta , Ratones , Plasmodium falciparum/genética , Plasmodium falciparum/inmunología
14.
Vaccine ; 15(8): 842-5, 1997 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-9234529

RESUMEN

Data generated in the Plasmodium yoelii rodent model indicated that plasmid DNA vaccines encoding the P.yoelii circumsporozoite protein (PyCSP) or 17 kDa hepatocyte erythrocyte protein (PyHEP17) were potent inducers of protective CD8+ T cell responses directed against infected hepatocytes. Immunization with a mixture of these plasmids circumvented the genetic restriction of protective immunity and induced additive protection. A third DNA vaccine encoding the P. yoelii sporozoite surface protein 2 (PySSP2) also induced protection. The P. falciparum genes encoding the homologues of these three protective P. yoelii antigens as well as another P. falciparum gene encoding a protein that is expressed in infected hepatocytes have been chosen for the development of a human vaccine. The optimal plasmid constructs for human use will be selected on the basis of immunogenicity data generated in mice and nonhuman primates. We anticipate that optimization of multi-gene P. falciparum DNA vaccines designed to protect against malaria by inducing CD8+ T cells that target infected hepatocytes will require extensive clinical trials during the coming years.


Asunto(s)
Vacunas contra la Malaria , Malaria Falciparum/prevención & control , Plasmodium falciparum/genética , Vacunas de ADN , Animales , Antígenos de Protozoos/genética , Antígenos de Protozoos/inmunología , ADN Protozoario/inmunología , Modelos Animales de Enfermedad , Eritrocitos/parasitología , Humanos , Malaria/inmunología , Malaria/prevención & control , Vacunas contra la Malaria/genética , Vacunas contra la Malaria/inmunología , Ratones , Ratones Endogámicos BALB C , Plasmodium falciparum/inmunología , Plasmodium yoelii/genética , Plasmodium yoelii/inmunología , Proteínas Protozoarias/genética , Proteínas Protozoarias/inmunología
16.
J Infect Dis ; 171(6): 1569-75, 1995 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-7769294

RESUMEN

Primaquine was tested as a prophylactic drug against Plasmodium falciparum in a region in western Kenya in which malaria is holoendemic. Children 9-14 years old were randomized to receive regimens of daily primaquine, daily doxycycline, daily proguanil plus weekly chloroquine, daily vitamin plus weekly mefloquine, or daily vitamin alone. Primaquine, doxycycline, and mefloquine were equally effective in preventing both symptomatic and asymptomatic malarial infections. Chloroquine plus proguanil was the least effective regimen. There was no toxicity from daily primaquine during the 11 weeks of the study. Findings show that primaquine can be successfully used as a causal prophylactic regimen against falciparum malaria in western Kenya; chloroquine plus proguanil was not as efficacious as the three other preventive regimens; most Kenyan children receiving standard doses of mefloquine and doxycycline had lower than expected serum trough drug levels; and some volunteers with adequate mefloquine or doxycycline levels at trough developed asymptomatic parasitemias and clinical malaria.


Asunto(s)
Malaria Falciparum/prevención & control , Primaquina/administración & dosificación , Adolescente , Niño , Cloroquina/administración & dosificación , Doxiciclina/administración & dosificación , Esquema de Medicación , Humanos , Kenia , Mefloquina/administración & dosificación , Proguanil/administración & dosificación
17.
Lancet ; 343(8897): 564-8, 1994 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-7906328

RESUMEN

Two field studies in Kenya and an experimental challenge study in the USA were done to assess the accuracy of a dipstick antigen-capture assay based on qualitative detection of Plasmodium falciparum histidine-rich protein 2 (PfHRP-2) in peripheral blood for diagnosis of P falciparum infection. In these studies, the assay was 96.5-100% sensitive for detection of greater than 60 P falciparum asexual parasites/microL blood, 70-81% sensitive for 11-60 parasites/microL blood, and 11-67% sensitive for 10 parasites or less/microL blood. Specificity was 95% (95% CI 85-105%; n = 20) among naive American volunteers, 98% (96-101%; n = 112) among volunteers exposed to the bite of P falciparum-infected mosquitoes, and 88% (84-92%; n = 285) among Kenyans living in an area with holoendemic malaria. Our results also indicated that PfHRP-2 antigen was not detectable in blood 6 days after initiation of curative chemotherapy, and suggest that such circulating antigens rarely lead to false-positive tests. The dipstick assay's sensitivity, specificity, simplicity, and speed may make it an important tool in the battle against malaria.


Asunto(s)
Antígenos de Protozoos/sangre , Malaria Falciparum/diagnóstico , Plasmodium falciparum/inmunología , Proteínas Protozoarias/sangre , Adolescente , Adulto , Animales , Niño , Femenino , Humanos , Malaria Falciparum/inmunología , Masculino , Valor Predictivo de las Pruebas , Tiras Reactivas , Sensibilidad y Especificidad
18.
J Immunol ; 151(5): 2690-8, 1993 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-8103069

RESUMEN

BALB/c mice are strongly protected against malaria after immunization with Plasmodium yoelii (PY) sporozoites, and this is an important model for malaria vaccine development in humans. This paper explores the role of CD4+ cells in the induction of the antimalarial immune response. The method used has been to treat animals with anti-CD4 mAb before immunization, resulting in a profound depletion of CD4+ T cells. The primary finding is that mice are not protected by sporozoite immunization after depletion of CD4+ cells. Such mice make little antibody to malaria sporozoite Ag, showing the strong T-cell dependence of these humoral responses. However, infusion of hyperimmune serum does not restore immunity to anti-CD4 treated animals. Neither does injection of exogenous IL-2 compensate for the absence of CD4+ cells. However, regrowth of CD4+ cells does allow successful immunization of animals, showing that long-term suppression against malaria Ag has not been induced by immunization in the absence of CD4+ cells. It is thought that infiltrating CD8+ T cells are critical effector cells against PY parasites in the pre-erythrocytic stages. Mice immunized while depleted of CD4+ cells have normal numbers of CD8+ T cells infiltrating their livers. In addition, they have normal numbers of splenic CD8+ CTL directed at the PY circumsporozoite protein. Thus, it appears that although CD8+ cells have been activated in the absence of CD4+ cells, they cannot protect mice against malaria. We conclude that a successful vaccine against the pre-erythrocytic stages of malaria must activate both CD4+ and CD8+ T cells.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Plasmodium yoelii/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/inmunología , Antígenos CD4/fisiología , Antígenos CD8/análisis , Células Cultivadas , Inmunización , Interleucina-2/farmacología , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Linfocitos T Citotóxicos/fisiología
19.
J Immunol ; 149(6): 2103-9, 1992 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-1517574

RESUMEN

Clone B is a cytotoxic T cell clone induced by immunization with Plasmodium yoelii sporozoites which recognizes an epitope on both the P. yoelii and Plasmodium berghei circumsporozoite proteins. It is CD8, uses the V beta 8.1 TCR, and is Kd restricted. When adoptively transferred, it protects mice against infection by both species of malaria sporozoites, and this protection is dependent on IFN-gamma. Clone B cells are more broadly reactive and protective than previously described murine T cell clones against malaria. Clone B may be an important model for immune protection against the spectrum of variant parasites in nature.


Asunto(s)
Antígenos de Protozoos/inmunología , Plasmodium berghei/inmunología , Plasmodium yoelii/inmunología , Proteínas Protozoarias/inmunología , Linfocitos T Citotóxicos/inmunología , Secuencia de Aminoácidos , Animales , Antígenos de Protozoos/química , Células Clonales , Reacciones Cruzadas , Inmunización Pasiva , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Péptidos/inmunología , Proteínas Protozoarias/química
20.
Vaccine ; 10(9): 578-84, 1992.
Artículo en Inglés | MEDLINE | ID: mdl-1323900

RESUMEN

In Balb/c mice, the sterile protective immunity induced by immunization with radiation-attenuated Plasmodium yoelii sporozoites is eliminated by in vivo depletion of CD8+ T lymphocytes, suggesting that cytotoxic T lymphocytes (CTL) against malaria antigens expressed on infected hepatocytes are required for mediating this protective immunity. To produce a vaccine that would induce CTL against the P. yoelii circumsporozoite protein (CS), we constructed an attenuated pseudorabies virus (PRV) containing a gene encoding this protein. Balb/c mice that received three doses of 10(7) plaque-forming units (p.f.u.) of this vaccine intravenously at 3 week intervals developed high levels of antibodies to sporozoites (indirect fluorescent antibody titre = 4096) and CTL against a 16 amino acid epitope (SYVPSAEQILEFVKQI, amino acids 281-296) from the P. yoelii CS protein designated PYCTL1. The cytotoxic activity of the CTL was antigen-specific, MHC-restricted, and dependent on CD8+ T cells. Furthermore, these CTL eliminated P. yoelii-infected hepatocytes from in vitro culture, indicating that they recognize this peptide on the surface of infected hepatocytes. However, all nine mice that were challenged with 200 sporozoites developed a blood-stage malaria infection. We attribute this lack of protection to the great difficulty of inducing sterile immunity against this highly infectious parasite P. yoelii. We conclude that recombinant pseudorabies virus (PRV) worked successfully as a live vaccine vector to induce both antibodies and CTL, albeit non-protective in vivo, and the herpesviruses should be considered as subunit vaccines where T- and B-cell immunity is required.


Asunto(s)
Antígenos de Protozoos/genética , Malaria/inmunología , Plasmodium yoelii/genética , Plasmodium yoelii/inmunología , Proteínas Protozoarias , Secuencia de Aminoácidos , Animales , Anticuerpos Antiprotozoarios/biosíntesis , Antígenos de Protozoos/inmunología , Linfocitos B/inmunología , Femenino , Vectores Genéticos/inmunología , Herpesvirus Suido 1/genética , Inmunidad , Inmunización , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Péptidos/genética , Péptidos/inmunología , Linfocitos T Citotóxicos/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...