Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Sci Rep ; 11(1): 21384, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34725414

RESUMEN

Group B Streptococcus (GBS) is generally an asymptomatic colonizer of human mucosa but it occasionally infects pregnant women and neonates through vertical transmission, causing disease during the first weeks of life with frequent and severe complications. Preclinical studies have shown that maternal vaccination with polysaccharide-based vaccines protects mothers and offspring from GBS mucosal colonization and consecutive infection. In these models, bacteria were inoculated in mouse either intravaginally in the last trimester of pregnancy or systemically in pups. Here, we investigated whether maternal vaccination with glycoconjugate vaccines may also prevent GBS-mediated colonization and disease in neonates using an infection route that more closely mimics inhalation or ingestion of bacteria during human delivery. To address this point, mice aged less than two days were intranasally challenged with epidemiologically relevant GBS strains. Bacteria were found to colonize nose and intestine, reaching in some cases lungs and blood during the first days of life. Bacteria were also found in vagina of a fraction of colonized female mice within the first month of life. GBS-specific IgG induced by maternal vaccination with a glycoconjugate vaccine formulation were found in blood and mucosal tissues of newborns. Finally, when intranasally challenged with GBS serotype III strains, pups delivered by vaccinated mothers were partially protected against mucosal colonization and deeper infection.


Asunto(s)
Glicoconjugados/uso terapéutico , Infecciones Estreptocócicas/prevención & control , Vacunas Estreptocócicas/uso terapéutico , Streptococcus agalactiae/inmunología , Animales , Animales Recién Nacidos , Femenino , Inmunidad Materno-Adquirida , Transmisión Vertical de Enfermedad Infecciosa/prevención & control , Ratones , Embarazo , Infecciones Estreptocócicas/inmunología
2.
J Immunol Methods ; 316(1-2): 84-96, 2006 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-17010367

RESUMEN

Among the experimental animal models, mice remain the most widely used for the evaluation of immunotherapeutic strategies. Vaccines against parasites and viral antigens are commonly administered to the appropriate mouse strain which also allows testing of the therapeutic effect. Similarly, in mice transgenic for human tumor associated antigens (TAA), cancer vaccines must lead to breakage of immune tolerance to elicit a significant effect on the tumor. However, one of the major drawbacks in the monitoring of cellular immune responses induced by vaccination is that functional immunological assays require suppression of the animals to collect the spleen or lymph nodes for analysis. Here, we report the application of a rapid intracellular staining (ICS) method to quantify antigen-specific T cells responses in small volumes of murine blood. Genetic vaccination with plasmid DNA followed by electroporation (DNA-EP) and the use of adenoviral vectors (Ad) encoding CEA as a model target antigen were applied to different strains of mice. Optimal blood volume, number of lymphocytes, sensitivity and reproducibility of intracellular staining for IFN-gamma were determined both in non-tolerant/wild type mice as well as in tolerant CEA transgenic mice upon restimulation of PBMCs with CEA peptides. Groups of vaccinated mice were then sacrificed and PBMCs and splenocytes from individual animals were compared for intracytoplasmic detection of IFN-gamma and TNF-alpha. A significant correlation was observed between splenic and blood immune responses. Finally, the cellular immune response was followed over time in groups of vaccinated mice. The kinetics of IFN-gamma producing effectors were measured after priming and successive boosting with adenoviral vectors. We show that intracellular staining for mouse PBMCs is a rapid and simple method to measure antigen-specific immune responses. It does not require animal euthanasia and mirrors the response observed in lymphoid organs such as the spleen.


Asunto(s)
Antígeno Carcinoembrionario/inmunología , Ratones/sangre , Coloración y Etiquetado/métodos , Linfocitos T/inmunología , Animales , Antígeno Carcinoembrionario/sangre , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Fluoresceína-5-Isotiocianato/química , Interferón gamma/sangre , Interleucina-2/sangre , Ratones/inmunología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Bazo/inmunología , Factor de Necrosis Tumoral alfa/sangre
3.
Sci Transl Med ; 3(91): 91ra62, 2011 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-21753121

RESUMEN

The sequence variability of protective antigens is a major challenge to the development of vaccines. For Neisseria meningitidis, the bacterial pathogen that causes meningitis, the amino acid sequence of the protective antigen factor H binding protein (fHBP) has more than 300 variations. These sequence differences can be classified into three distinct groups of antigenic variants that do not induce cross-protective immunity. Our goal was to generate a single antigen that would induce immunity against all known sequence variants of N. meningitidis. To achieve this, we rationally designed, expressed, and purified 54 different mutants of fHBP and tested them in mice for the induction of protective immunity. We identified and determined the crystal structure of a lead chimeric antigen that was able to induce high levels of cross-protective antibodies in mice against all variant strains tested. The new fHBP antigen had a conserved backbone that carried an engineered surface containing specificities for all three variant groups. We demonstrate that the structure-based design of multiple immunodominant antigenic surfaces on a single protein scaffold is possible and represents an effective way to create broadly protective vaccines.


Asunto(s)
Antígenos Bacterianos/inmunología , Diseño de Fármacos , Inmunidad/inmunología , Neisseria meningitidis/inmunología , Animales , Antibacterianos/farmacología , Antígenos Bacterianos/química , Proteínas Bacterianas/química , Proteínas Bacterianas/inmunología , Cristalografía por Rayos X , Humanos , Inmunidad/efectos de los fármacos , Espectroscopía de Resonancia Magnética , Ratones , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/inmunología , Mutación/genética , Neisseria meningitidis/efectos de los fármacos , Ingeniería de Proteínas , Estructura Secundaria de Proteína
4.
J Immunother ; 32(7): 744-54, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19561534

RESUMEN

Human leukocyte antigen (HLA)-A2.1 transgenic mice (HHD) represent a valuable model to study and predict the immunogenicity of vaccines against pathogens. However, HHD mice are unsuitable for in vivo studies of cancer vaccines against human tumor-associated antigens because they lack T-cell tolerance that is key to define the potency of the treatment. In this study, we developed HHD/carcinoembryonic antigen P(CEA) hybrid mice by breeding transgenic mice homozygous for CEA with HHD. These mice express human CEA, present epitopes solely through HLA-A2.1 molecules and constitute a unique in vivo animal model to study HLA-A2.1-restricted immune response of a human CEA-based vaccine. Owing to the immune tolerance, HHD/CEA mice show a limited immune response and expansion of a different and restricted T-cell receptor repertoire after antigen-specific stimulation. Our data show that genetic vectors expressing CEA and peptide-based vaccines are able to efficiently break immune tolerance against CEA and to elicit strong immune response against HLA-A2.1-restricted CEA epitopes. Most importantly, efficient lysis of human CEA+/HLA-A2.1+ tumor cells was observed and significant protection against HHD/CEA tumor cells was achieved in HHD/CEA-vaccinated mice. Hence, HHD/CEA provides a relevant model for the evaluation of the potential efficacy of human CEA-based vaccines.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Antígeno Carcinoembrionario/inmunología , Antígeno HLA-A2/inmunología , Melanoma Experimental/inmunología , Adenoviridae/genética , Animales , Vacunas contra el Cáncer/administración & dosificación , Antígeno Carcinoembrionario/genética , Antígeno Carcinoembrionario/metabolismo , Línea Celular Tumoral , Citotoxicidad Inmunológica/inmunología , Ensayo de Inmunoadsorción Enzimática , Epítopos/inmunología , Heces/química , Femenino , Citometría de Flujo , Tracto Gastrointestinal/metabolismo , Antígenos H-2/inmunología , Antígeno HLA-A2/genética , Antígeno HLA-A2/metabolismo , Humanos , Masculino , Melanoma Experimental/patología , Melanoma Experimental/prevención & control , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Transfección , Vacunación/métodos
5.
Hum Gene Ther ; 20(2): 125-36, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18937552

RESUMEN

A genetic vaccine platform based on DNA electroporation (DNA-EP) and adenovirus (Ad) was used to generate immune response against human carcinoembryonic antigen (CEA) and antitumor effects in murine models with spontaneous tumors arising in an orthotopic location. CEA transgenic (CEA.Tg) mice treated with the carcinogen 1,2-dimethylhydrazine developed CEA-overexpressing tumors that resembled human sporadic colorectal cancer. APC1638N/CEA hybrid mice, generated by crossing mice carrying the adenomatous polyposis coli (Apc1638N) gene mutation with CEA.Tg mice, are representative of human familial polyposis and develop polyps that overexpress the antigen. In both models, the DNA-EP/Ad vaccine succeeded in breaking immune tolerance and achieved significant antitumor effects in therapeutic settings. Our data suggest that genetic vaccines targeting CEA may be feasible strategies against gut tumors that overexpress the antigen. In addition, these models are powerful systems for evaluating antigen-specific tumor immunity and assessing therapeutic vaccine strategies for human colorectal cancer.


Asunto(s)
Pólipos Adenomatosos/terapia , Vacunas contra el Cáncer/uso terapéutico , Antígeno Carcinoembrionario/metabolismo , Neoplasias Colorrectales/terapia , Vectores Genéticos , Pólipos Adenomatosos/inmunología , Pólipos Adenomatosos/patología , Adenoviridae/genética , Animales , Vacunas contra el Cáncer/genética , Antígeno Carcinoembrionario/genética , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , Modelos Animales de Enfermedad , Vectores Genéticos/genética , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Estándares de Referencia , Regulación hacia Arriba
6.
Int J Cancer ; 120(11): 2290-300, 2007 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-17304509

RESUMEN

Scaling up experimental protocols from rodents to humans is often not a straightforward procedure, and this particularly applies to cancer vaccines, where vaccination technology must be especially effective to overcome a variety of immune suppressive mechanisms. DNA electroporation (DNA-EP) and adenoviral vectors (Ad) have shown high potency and therapeutic efficacy for different antigens in several pre-clinical models. To evaluate the ability of DNA-EP and Ad to break tolerance to a self-antigen in large animals, we have cloned the CEA homologue (rhCEA) from rhesus monkeys (Macaca mulatta) colon tissue samples. rhCEA is a 705 aa protein and shares 78.9% homology to human CEA protein. Immunogenicity of rhCEA expressing vectors was tested in mice and subsequently in rhesus monkeys. To further increase the immunogenic potency of these vectors, a synthetic codon optimized rhCEA cDNA (rhCEAopt) was constructed. Genetic vaccination of rhesus monkeys was effective in breaking immune tolerance to rhCEA in all immunized animals, maintaining over time the elicited immune response, and most importantly, neither autoimmunity nor other side-effects were observed upon treatment. Our data confirm the efficacy of genetic cancer vaccines in large animals such as nonhuman primates and show that development of modified expression cassettes that result in increased potency of plasmid DNA and adenovirus may have a significant impact on vaccine development against malignancies expressing tumor associated antigens in patients.


Asunto(s)
Adenoviridae/inmunología , Vacunas contra el Cáncer/inmunología , Antígeno Carcinoembrionario/inmunología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Electroporación , Macaca mulatta , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido , Homología de Secuencia de Ácido Nucleico
7.
J Gene Med ; 7(8): 1086-96, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15772935

RESUMEN

BACKGROUND: Helper-dependent adenoviral (HD-Ad) vectors give rise to sustained gene expression after delivery in a variety of organisms. In particular, we previously documented persistent expression of erythropoietin (EPO) in mice after a single intramuscular (i.m.) injection of a HD-Ad vector harboring the mouse EPO cDNA. METHODS: We use the same vector harboring the tetracycline (tet)-dependent transactivator (rtTA2S-M2) and silencer (tTS) and mouse EPO cDNA to analyze the capacity of the dual tet-dependent transactivator system to control long-term EPO gene expression and to study the effect of an eventual immune response against these artificial proteins after i.m. delivery in immuno-competent mice. RESULTS: In the present study we demonstrate that i.m. injection of this vector in immuno-competent mice generates a cellular immune response to the rtTA2S-M2 transcription factor. This response curtails the duration of mouse EPO expression in mice, presumably by destroying the cells that co-express transcription factors and the therapeutic gene. Nonetheless, regulation of mouse EPO secretion was maintained during the entire experimental period, both when the vector dosage was reduced and when the tet-dependent transcription factors were put under the control of a muscle-specific promoter. CONCLUSIONS: Delivery of the tet transactivators using as vehicle a HD-Ad vector induced an immune response directed against the transactivators themselves, causing short-term therapeutic transgene expression. Regulated, long-term therapeutic transgene expression was, however, obtained by reducing the vector dose or expressing the transactivators under the control of a muscle-specific promoter.


Asunto(s)
Adenoviridae/genética , Eritropoyetina/genética , Eritropoyetina/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Virus Helper/fisiología , Tetraciclina/farmacología , Transactivadores/inmunología , Transactivadores/metabolismo , Animales , Electroporación , Femenino , Regulación de la Expresión Génica/inmunología , Vectores Genéticos/genética , Inmunidad Celular/inmunología , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/inmunología , Músculo Esquelético/metabolismo , Factores de Tiempo , Transactivadores/genética , Transgenes/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA