Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
PLoS Pathog ; 15(4): e1007721, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-31009499

RESUMEN

The need for improved dengue vaccines remains since the only licensed vaccine, Dengvaxia, shows variable efficacy depending on the infecting dengue virus (DENV) type, and increases the risk of hospitalization for severe dengue in children not exposed to DENV before vaccination. Here, we developed a tetravalent dengue purified and inactivated vaccine (DPIV) candidate and characterized, in rhesus macaques, its immunogenicity and efficacy to control DENV infection by analyzing, after challenge, both viral replication and changes in biological markers associated with dengue in humans. Although DPIV elicited cross-type and long-lasting DENV-neutralizing antibody responses, it failed to control DENV infection. Increased levels of viremia/RNAemia (correlating with serum capacity at enhancing DENV infection in vitro), AST, IL-10, IL-18 and IFN-γ, and decreased levels of IL-12 were detected in some vaccinated compared to non-vaccinated monkeys, indicating the vaccination may have triggered antibody-dependent enhancement of DENV infection. The dengue macaque model has been considered imperfect due to the lack of DENV-associated clinical signs. However, here we show that post-vaccination enhanced DENV infection can be detected in this model when integrating several parameters, including characterization of DENV-enhancing antibodies, viremia/RNAemia, and biomarkers relevant to dengue in humans. This improved dengue macaque model may be crucial for early assessment of efficacy and safety of future dengue vaccines.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Vacunas contra el Dengue/inmunología , Virus del Dengue/inmunología , Dengue/inmunología , Vacunas de Productos Inactivados/inmunología , Viremia/inmunología , Animales , Acrecentamiento Dependiente de Anticuerpo , Dengue/prevención & control , Dengue/virología , Vacunas contra el Dengue/administración & dosificación , Modelos Animales de Enfermedad , Femenino , Macaca mulatta , Masculino , Vacunación , Viremia/virología
2.
Tuberculosis (Edinb) ; 143: 102425, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-38180028

RESUMEN

A new efficacious tuberculosis vaccine targeting adolescents/adults represents an urgent medical need. The M72/AS01E vaccine candidate protected half of the latently-infected adults against progression to pulmonary tuberculosis in a Phase IIb trial (NCT01755598). We report that three immunizations of mice, two weeks apart, with AS01-adjuvanted M72 induced polyfunctional, Th1-cytokine-expressing M72-specific CD4+/CD8+ T cells in blood and lungs, with the highest frequencies in lungs. Antigen-dose reductions across the three vaccinations skewed pulmonary CD4+ T-cell profiles towards IL-17 expression. In blood, reducing antigen and adjuvant doses of only the third injection (to 1/5th or 1/25th of those of the first injections) did not significantly alter CD4+ T-cell/antibody responses; applying a 10-week delay for the fractional third dose enhanced antibody titers. Delaying a full-dose booster enhanced systemic CD4+ T-cell and antibody responses. Cross-reactivity with PPE and non-PPE proteins was assessed, as Mycobacterium tuberculosis (Mtb) virulence factors and evasion mechanisms are often associated with PE/PPE proteins, to which Mtb39a (contained in M72) belongs. In silico/in vivo analyses revealed that M72/AS01 induced cross-reactive systemic CD4+ T-cell responses to epitopes in a non-vaccine antigen (putative latency-associated Mtb protein PPE24/Rv1753c). These preclinical data describing novel mechanisms of M72/AS01-induced immunity could guide future clinical development of the vaccine.


Asunto(s)
Mycobacterium tuberculosis , Vacunas contra la Tuberculosis , Animales , Ratones , Linfocitos T CD8-positivos , Vacunación , Inmunización
3.
Sci Transl Med ; 15(710): eadg6050, 2023 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-37611082

RESUMEN

The RSVPreF3-AS01 vaccine, containing the respiratory syncytial virus (RSV) prefusion F protein and the AS01 adjuvant, was previously shown to boost neutralization responses against historical RSV strains and to be efficacious in preventing RSV-associated lower respiratory tract diseases in older adults. Although RSV F is highly conserved, variation does exist between strains. Here, we characterized variations in the major viral antigenic sites among contemporary RSV sequences when compared with RSVPreF3 and showed that, in older adults, RSVPreF3-AS01 broadly boosts neutralization responses against currently dominant and antigenically distant RSV strains. RSV-neutralizing responses are thought to play a central role in preventing RSV infection. Therefore, the breadth of RSVPreF3-AS01-elicited neutralization responses may contribute to vaccine efficacy against contemporary RSV strains and those that may emerge in the future.


Asunto(s)
Infecciones por Virus Sincitial Respiratorio , Vacunas , Humanos , Anciano , Virus Sincitiales Respiratorios , Infecciones por Virus Sincitial Respiratorio/prevención & control , Antígenos Virales
4.
PLoS Negl Trop Dis ; 14(4): e0008191, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32267846

RESUMEN

Dengue virus (DENV) is transmitted by infectious mosquitoes during blood-feeding via saliva containing biologically-active proteins. Here, we examined the effect of varying DENV infection modality in rhesus macaques in order to improve the DENV nonhuman primate (NHP) challenge model. NHPs were exposed to DENV-1 via subcutaneous or intradermal inoculation of virus only, intradermal inoculation of virus and salivary gland extract, or infectious mosquito feeding. The infectious mosquito feeding group exhibited delayed onset of viremia, greater viral loads, and altered clinical and immune responses compared to other groups. After 15 months, NHPs in the subcutaneous and infectious mosquito feeding groups were re-exposed to either DENV-1 or DENV-2. Viral replication and neutralizing antibody following homologous challenge were suggestive of sterilizing immunity, whereas heterologous challenge resulted in productive, yet reduced, DENV-2 replication and boosted neutralizing antibody. These results show that a more transmission-relevant exposure modality resulted in viral replication closer to that observed in humans.


Asunto(s)
Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Dengue/inmunología , Animales , Dengue/virología , Virus del Dengue/fisiología , Modelos Animales de Enfermedad , Femenino , Cinética , Macaca mulatta/inmunología , Mosquitos Vectores/virología , ARN Viral/sangre , Glándulas Salivales/virología , Vacunación , Carga Viral , Viremia/prevención & control , Replicación Viral
5.
PLoS One ; 13(4): e0196311, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29694440

RESUMEN

The macaque is widely accepted as a suitable model for preclinical characterization of dengue vaccine candidates. However, the only vaccine for which both preclinical and clinical efficacy results were reported so far showed efficacy levels that were substantially different between macaques and humans. We hypothesized that this model's predictive capacity may be improved using recent and minimally passaged dengue virus isolates, and by assessing vaccine efficacy by characterizing not only the post-dengue virus challenge viremia/RNAemia but also the associated-cytokine profile. Ten recent and minimally passaged Brazilian clinical isolates from the four dengue virus serotypes were tested for their infectivity in rhesus macaques. For the strains showing robust replication capacity, the associated-changes in soluble mediator levels, and the elicited dengue virus-neutralizing antibody responses, were also characterized. Three isolates from dengue virus serotypes 1, 2 and 4 induced viremia of high magnitude and longer duration relative to previously reported viremia kinetics in this model, and robust dengue virus-neutralizing antibody responses. Consistent with observations in humans, increased MCP-1, IFN-γ and VEGF-A levels, and transiently decreased IL-8 levels were detected after infection with the selected isolates. These results may contribute to establishing a dengue macaque model showing a higher predictability for vaccine efficacy in humans.


Asunto(s)
Virus del Dengue/inmunología , Dengue/patología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Brasil , Quimiocina CCL2/metabolismo , Chlorocebus aethiops , Dengue/veterinaria , Virus del Dengue/aislamiento & purificación , Regulación hacia Abajo , Interferón gamma/metabolismo , Interleucina-8 , Macaca mulatta , Serogrupo , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/metabolismo , Células Vero
6.
PLoS Negl Trop Dis ; 10(2): e0004423, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26919472

RESUMEN

BACKGROUND: A vaccine to prevent infection and disease caused by Plasmodium vivax is needed both to reduce the morbidity caused by this parasite and as a key component in efforts to eradicate malaria worldwide. Vivax malaria protein 1 (VMP001), a novel chimeric protein that incorporates the amino- and carboxy- terminal regions of the circumsporozoite protein (CSP) and a truncated repeat region that contains repeat sequences from both the VK210 (type 1) and the VK247 (type 2) parasites, was developed as a vaccine candidate for global use. METHODS: We conducted a first-in-human Phase 1 dose escalation vaccine study with controlled human malaria infection (CHMI) of VMP001 formulated in the GSK Adjuvant System AS01B. A total of 30 volunteers divided into 3 groups (10 per group) were given 3 intramuscular injections of 15 µg, 30 µg, or 60 µg respectively of VMP001, all formulated in 500 µL of AS01B at each immunization. All vaccinated volunteers participated in a P. vivax CHMI 14 days following the third immunization. Six non-vaccinated subjects served as infectivity controls. RESULTS: The vaccine was shown to be well tolerated and immunogenic. All volunteers generated robust humoral and cellular immune responses to the vaccine antigen. Vaccination did not induce sterile protection; however, a small but significant delay in time to parasitemia was seen in 59% of vaccinated subjects compared to the control group. An association was identified between levels of anti-type 1 repeat antibodies and prepatent period. SIGNIFICANCE: This trial was the first to assess the efficacy of a P. vivax CSP vaccine candidate by CHMI. The association of type 1 repeat-specific antibody responses with delay in the prepatency period suggests that augmenting the immune responses to this domain may improve strain-specific vaccine efficacy. The availability of a P. vivax CHMI model will accelerate the process of P. vivax vaccine development, allowing better selection of candidate vaccines for advancement to field trials.


Asunto(s)
Vacunas contra la Malaria/inmunología , Malaria Vivax/prevención & control , Plasmodium vivax/inmunología , Proteínas Protozoarias/inmunología , Adolescente , Adulto , Anticuerpos Antiprotozoarios/inmunología , Femenino , Humanos , Vacunas contra la Malaria/administración & dosificación , Vacunas contra la Malaria/efectos adversos , Malaria Vivax/inmunología , Malaria Vivax/parasitología , Masculino , Persona de Mediana Edad , Proteínas Protozoarias/administración & dosificación , Proteínas Protozoarias/efectos adversos , Vacunación , Adulto Joven
7.
J Vet Diagn Invest ; 17(3): 295-8, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15945393

RESUMEN

The purpose of this study was to characterize the incidence and diagnostic features of amyloidosis and other diseases found at necropsy in captive trumpeter swans (Cygnus buccinator). A search of Iowa State University's Department of Veterinary Pathology and Veterinary Diagnostic Laboratory databases yielded 31 trumpeter swan (C. buccinator) necropsy cases from captive swans in protected habitats. Eleven of the 31 birds had amyloid deposition most commonly in the spleen (8 of 11), liver (7 of 11), and kidney (6 of 11) and less often in the pancreas (2 of 11) and adrenal gland (2 of 11). Amyloid deposition effaced normal tissue with adjacent necrosis and hemorrhage in severe cases. Amyloidosis was most often diagnosed in February and March. Other disease diagnoses in the trumpeter swans included aspergillosis (5 of 31, 16%); bacterial infection (5 of 31, 16%); lead toxicosis (3 of 31, 10%); gout (2 of 31, 6%); parasitic infection (2 of 31, 6%); vitamin E deficiency (1 of 31, 3%); trauma (1 of 31, 3%); and ventricular foreign body (1 of 31, 3%). Histopathologic, toxicologic, and microbiologic analyses did not define an etiologic diagnosis in the deaths of 9 trumpeter swans. In these cases, necropsy lesions included emaciation (5 of 9), enteritis (1 of 9), pulmonary hemorrhage (1 of 9), and no lesions (3 of 9). The number of trumpeter swan case submissions was greatest in January and February. This study provides a reference for veterinary diagnosticians concerning incidence and diagnostic features of amyloidosis and other diseases in captive trumpeter swans of the midwestern United States.


Asunto(s)
Amiloidosis/veterinaria , Anseriformes , Enfermedades de las Aves/epidemiología , Amiloidosis/epidemiología , Animales , Femenino , Cuerpos Extraños/epidemiología , Cuerpos Extraños/veterinaria , Infecciones/epidemiología , Infecciones/veterinaria , Intoxicación por Plomo/epidemiología , Intoxicación por Plomo/veterinaria , Masculino , Estudios Retrospectivos
8.
PLoS One ; 10(4): e0122835, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25856308

RESUMEN

HIV-1-specific CD4+ and CD8+ T lymphocytes are important for HIV-1 replication control. F4/AS01 consists of F4 recombinant fusion protein (containing clade B Gag/p24, Pol/RT, Nef and Gag/p17) formulated in AS01 Adjuvant System, and was shown to induce F4-specific polyfunctional CD4+ T-cell responses in humans. While replication-incompetent recombinant HIV-1/SIV antigen-expressing human adenoviral vectors can elicit high-frequency antigen-specific CD8+ T-cell responses, their use is hampered by widespread pre-existing immunity to human serotypes. Non-human adenovirus serotypes associated with lower prevalence may offer an alternative strategy. We evaluated the immunogenicity of AdC7-GRN ('A'), a recombinant chimpanzee adenovirus type 7 vector expressing clade B Gag, RT and Nef, and F4/AS01 ('P'), when delivered intramuscularly in homologous (PP or AA) and heterologous (AAPP or PPAA) prime-boost regimens, in macaques and mice. Vaccine-induced HIV-1-antigen-specific T cells in peripheral blood (macaques), liver, spleen, and intestinal and genital mucosa (mice) were characterized by intracellular cytokine staining. Vaccine-specific IgG antibodies (macaques) were detected using ELISA. In macaques, only the heterologous prime-boost regimens induced polyfunctional, persistent and balanced CD4+ and CD8+ T-cell responses specific to each HIV-1 vaccine antigen. AdC7-GRN priming increased the polyfunctionality of F4/AS01-induced CD4+ T cells. Approximately 50% of AdC7-GRN-induced memory CD8+ T cells exhibited an effector-memory phenotype. HIV-1-specific antibodies were detected with each regimen. In mice, antigen-specific CD4+ and CD8+ T-cell responses were detected in the mucosal and systemic anatomical compartments assessed. When administered in heterologous prime-boost regimens, AdC7-GRN and F4/AS01 candidate vaccines acted complementarily in inducing potent and persistent peripheral blood HIV-1-specific CD4+ and CD8+ T-cell responses and antibodies in macaques. Besides, adenoviral vector priming modulated the cytokine-expression profile of the protein-induced CD4+ T cells. Each regimen induced HIV-1-specific T-cell responses in systemic/local tissues in mice. This suggests that prime-boost regimens combining adjuvanted protein and low-seroprevalent chimpanzee adenoviral vectors represent an attractive vaccination strategy for clinical evaluation.


Asunto(s)
Vectores Genéticos/genética , VIH-1/inmunología , Proteínas Recombinantes de Fusión/inmunología , Linfocitos T/inmunología , Vacunas Virales/inmunología , Adenoviridae , Animales , Anticuerpos Antivirales/sangre , Citocinas/inmunología , Ensayo de Inmunoadsorción Enzimática , Genes pol/genética , Antígenos VIH/genética , Proteína p24 del Núcleo del VIH/genética , Inyecciones Intramusculares , Macaca , Ratones , Pan troglodytes , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/genética , Vacunas Virales/administración & dosificación , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/genética
9.
PLoS One ; 10(7): e0131571, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26148007

RESUMEN

METHODS: In an observer blind, phase 2 trial, 55 adults were randomized to receive one dose of Ad35.CS.01 vaccine followed by two doses of RTS,S/AS01 (ARR-group) or three doses of RTS,S/AS01 (RRR-group) at months 0, 1, 2 followed by controlled human malaria infection. RESULTS: ARR and RRR vaccine regimens were well tolerated. Efficacy of ARR and RRR groups after controlled human malaria infection was 44% (95% confidence interval 21%-60%) and 52% (25%-70%), respectively. The RRR-group had greater anti-CS specific IgG titers than did the ARR-group. There were higher numbers of CS-specific CD4 T-cells expressing > 2 cytokine/activation markers and more ex vivo IFN-γ enzyme-linked immunospots in the ARR-group than the RRR-group. Protected subjects had higher CS-specific IgG titers than non-protected subjects (geometric mean titer, 120.8 vs 51.8 EU/ml, respectively; P = .001). CONCLUSIONS: An increase in vaccine efficacy of ARR-group over RRR-group was not achieved. Future strategies to improve upon RTS,S-induced protection may need to utilize alternative highly immunogenic prime-boost regimens and/or additional target antigens. TRIAL REGISTRATION: ClinicalTrials.gov NCT01366534.


Asunto(s)
Vacunas contra la Malaria/inmunología , Malaria/inmunología , Malaria/prevención & control , Esporozoítos/inmunología , Anticuerpos Antiprotozoarios/inmunología , Formación de Anticuerpos/inmunología , Linfocitos T CD4-Positivos/inmunología , Método Doble Ciego , Humanos , Inmunización Secundaria/métodos , Inmunoglobulina G/inmunología , Pruebas Inmunológicas/métodos , Interferón gamma/inmunología , Vacunación/métodos
10.
Ann N Y Acad Sci ; 1026: 267-72, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15604504

RESUMEN

Leishmania parasites are vector-borne protozoal pathogens found in tropical and subtropical regions of both the Old and New World. These parasites can cause visceral or cutaneous disease, and the pathology of the infection is determined by both host immune factors and species/strain differences of the parasite. Dogs are an important reservoir for maintaining the population of Leishmania parasites that can lead to visceral leishmaniasis in humans, and a vaccination approach may be an effective method for reducing the numbers of infected dogs. Resistance to leishmaniasis has been consistently associated with a T helper 1 immune response, characterized by the production of IFN-gamma by the antigen-specific lymphocyte population. The development of this Th1 response has been shown to be dependent upon both cytokines and dendritic cells during T cell activation. However, the development of a Leishmania vaccine effective in preventing these chronic diseases has proven to be a challenge. Vaccine trials have focused on whole-killed or subunit vaccines with adjuvants. Newer experimental strategies involve the attenuation of the Leishmania parasite via gene deletion technologies or the expression of specific Leishmania peptides within attenuated organisms, such as Bacillus Calmette Guérin. DNA vaccines and dendritic cell potentiators, such as CpG oligodeoxynucleotides and Flt-3 ligand, are also in the early stage of development. In addition, as part of blocking the transmission cycle of leishmaniasis, several laboratories are also exploring the possibility of immunomodulating the host toward the bite of the sand fly.


Asunto(s)
Leishmania/inmunología , Leishmania/patogenicidad , Leishmaniasis/inmunología , Leishmaniasis/prevención & control , Vacunas Antiprotozoos , Animales , Enfermedad Crónica , Ensayos Clínicos como Asunto , Células Dendríticas , Reservorios de Enfermedades , Perros , Humanos , Linfocitos T Colaboradores-Inductores/inmunología , Vacunas de ADN
11.
Am J Trop Med Hyg ; 71(4): 407-11, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15516635

RESUMEN

The Th1 response elicited in mice infected with Leishmania major has been used as a model to characterize cellular immune defects associated with L. amazonensis infection. However, it is not known if the immune response associated with the infection by virulent L. major parasites can promote resistance to a subsequent L. amazonensis infection. Our data demonstrate that C3HeB/FeJ mice infected subcutaneously with virulent L. major are resistant to an L. amazonensis challenge. The healing phenotype is characterized by a Th1 response as measured by increased production of interferon-gamma and low levels of interleukin-4 in the draining lymph node. Together, this indicates that the Th1 response associated with L. major infection can promote resistance to L. amazonensis infection and that it can be used as a tool to study the immune defects associated with L. amazonensis infection.


Asunto(s)
Leishmania major/inmunología , Leishmania mexicana/inmunología , Leishmania mexicana/patogenicidad , Leishmaniasis Cutánea/inmunología , Células TH1/inmunología , Animales , Modelos Animales de Enfermedad , Humanos , Interferón gamma/metabolismo , Interleucina-4/metabolismo , Leishmaniasis Cutánea/parasitología , Ganglios Linfáticos/inmunología , Ratones , Ratones Endogámicos C3H
12.
Vaccine ; 31(52): 6216-24, 2013 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-24144477

RESUMEN

We have designed a pre-erythrocytic vaccine candidate based on the Plasmodium vivax circumsporozoite (CSV) protein, which includes its N- and C-terminal parts and a truncated region containing repeat sequences from both the VK210 and the VK247 P. vivax subtypes. Two versions of this vaccine candidate were made: a soluble recombinant protein expressed in Escherichia coli, designated VMP001 and a particulate antigen expressed in Saccharomyces cerevisiae, designated CSV-S,S. The latter is composed of CSV-S, a fusion protein between VMP001 and hepatitis B surface antigen (HBsAg), and free HBsAg co-expressed in yeast and self-assembling into mixed particles. Both antigen versions, adjuvanted with AS01, were shown to be immunogenic in rhesus monkeys. CSV-S,S/AS01 induced higher levels of VMP001-specific antibodies than did VMP001/AS01. Antibody responses against the N- and C-terminal regions of CSV and the VK210 repeat motif were of a similar magnitude following immunization with either the soluble or the particulate antigen. However, antibodies against the AGDR region, a potentially protective B cell epitope, were only detected after immunization with CSV-S,S. Analysis of the induced CD4(+) T cells highlighted different cytokine profiles depending on the antigen form. These results warrant further clinical evaluation of these two vaccine candidates to assess the added value of a particulate versus soluble form of CSV, in terms of both immunogenicity and protective efficacy.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Vacunas contra la Malaria/inmunología , Plasmodium vivax/inmunología , Proteínas Protozoarias/inmunología , Animales , Anticuerpos Antiprotozoarios/sangre , Linfocitos T CD4-Positivos/inmunología , Citocinas/metabolismo , Escherichia coli/genética , Expresión Génica , Macaca mulatta , Vacunas contra la Malaria/administración & dosificación , Vacunas contra la Malaria/genética , Plasmodium vivax/genética , Proteínas Protozoarias/genética , Saccharomyces cerevisiae/genética , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/genética , Vacunas de Subunidad/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Vacunas de Partículas Similares a Virus/administración & dosificación , Vacunas de Partículas Similares a Virus/genética , Vacunas de Partículas Similares a Virus/inmunología
13.
PLoS One ; 6(4): e19376, 2011 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-21559416

RESUMEN

During the course of a microbial infection, different antigen presenting cells (APCs) are exposed and contribute to the ensuing immune response. CD8α(+) dendritic cells (DCs) are an important coordinator of early immune responses to the intracellular bacteria Listeria monocytogenes (Lm) and are crucial for CD8(+) T cell immunity. In this study, we examine the contribution of different primary APCs to inducing immune responses against Lm. We find that CD8α(+) DCs are the most susceptible to infection while plasmacytoid DCs are not infected. Moreover, CD8α(+) DCs are the only DC subset capable of priming an immune response to Lm in vitro and are also the only APC studied that do so when transferred into ß2 microglobulin deficient mice which lack endogenous cross-presentation. Upon infection, CD11b(+) DCs primarily secrete low levels of TNFα while CD8α(+) DCs secrete IL-12 p70. Infected monocytes secrete high levels of TNFα and IL-12p70, cytokines associated with activated inflammatory macrophages. Furthermore, co-culture of infected CD8α(+) DCs and CD11b+ DCs with monocytes enhances production of IL-12 p70 and TNFα. However, the presence of monocytes in DC/T cell co-cultures attenuates T cell priming against Lm-derived antigens in vitro and in vivo. This suppressive activity of spleen-derived monocytes is mediated in part by both TNFα and inducible nitric oxide synthase (iNOS). Thus these monocytes enhance IL-12 production to Lm infection, but concurrently abrogate DC-mediated T cell priming.


Asunto(s)
Antígenos CD8/biosíntesis , Linfocitos T CD8-positivos/citología , Células Dendríticas/citología , Listeria monocytogenes/metabolismo , Listeriosis/sangre , Animales , Linfocitos T CD8-positivos/microbiología , Células Dendríticas/microbiología , Hibridomas/metabolismo , Interferón gamma/metabolismo , Interleucina-12/metabolismo , Melanoma Experimental , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Óxido Nítrico Sintasa de Tipo II/metabolismo , Péptidos/química , Factor de Necrosis Tumoral alfa/metabolismo
14.
Vaccine ; 29(43): 7335-42, 2011 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-21807053

RESUMEN

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


Asunto(s)
Antígenos de Protozoos/inmunología , Epítopos de Linfocito T/inmunología , Vacunas contra la Malaria/inmunología , Plasmodium yoelii/inmunología , Proteínas Protozoarias/inmunología , Esporozoítos/inmunología , Adenoviridae/genética , Adenoviridae/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Femenino , Inmunización , Interferón gamma , Malaria/inmunología , Malaria/prevención & control , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Esporozoítos/efectos de la radiación
15.
Infect Immun ; 74(3): 1547-54, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16495525

RESUMEN

C3HeB/FeJ mice challenged with Leishmania major develop a polarized Th1 response and subsequently heal, whereas Leishmania amazonensis challenge leads to chronic lesions with high parasite loads at 10 weeks postinfection. In this study, a comparison of draining lymph node cells from L. amazonensis- and L. major-infected mice at 10 weeks postinfection showed equivalent percentages of effector/memory phenotype CD44hi CD4+ T cells producing interleukin-2 (IL-2) and proliferating after antigen stimulation. However, these cells isolated from L. amazonensis-infected mice were not skewed toward either a Th1 or Th2 phenotype in vivo, as evidenced by their unbiased Th1/Th2 transcription factor mRNA profile. In vivo antigen stimulation with added IL-12 failed to enhance gamma interferon (IFN-gamma) production of CD4+ T cells from L. amazonensis-infected mice. Antigen stimulation of CD4+ T cells from L. amazonensis-infected mice in vitro in the presence of IL-12 resulted in production of only 10 to 15% of the IFN-gamma produced by T cells from L. major-infected mice under identical conditions. These results suggest that the CD4+ T-cell response during chronic L. amazonensis infection is limited during the transition from an early activated CD4+ T-cell population to an effector cell population and demonstrate that these T cells have an intrinsic defect beyond the presence or absence of IL-12 during antigen stimulation.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Leishmania , Leishmaniasis Cutánea/inmunología , Animales , Antígenos CD4/inmunología , Enfermedad Crónica , Interleucina-12/metabolismo , Leishmania/química , Ratones , Ratones Endogámicos C3H , Fenotipo
16.
Infect Immun ; 72(8): 4455-63, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15271903

RESUMEN

The susceptibility of mice to Leishmania amazonensis infection is thought to result from an inability to develop a Th1 response. Our data show that the low levels of gamma interferon (IFN-gamma) produced by the draining lymph node (DLN) cells of chronically infected mice could be enhanced in vitro and in vivo with L. amazonensis antigen-pulsed bone marrow-derived dendritic cells (BM-DC) and the Th1-promoting cytokine interleukin-12 (IL-12). Given intralesionally to chronically infected mice, this treatment induced the upregulation of mRNA levels for IFN-gamma, the transcription factor T-box expressed in T cells, and IL-12 receptor beta 2 in CD4(+) T cells from the DLN and an increase in parasite-specific immunoglobulin G2a in the serum. However, this Th1 response was not associated with healing, and the antigen-specific enhancement of IFN-gamma production remained impaired in the DLN. However, addition of IL-12 to the in vitro recall response was able to recover this defect, suggesting that antigen-presenting cell-derived IL-12 production may be limited in infected mice. This was supported by the fact that L. amazonensis amastigotes limited the production of IL-12p40 from BM-DC in vitro. Altogether, our data indicate that the immune response of mice chronically infected with L. amazonensis can be enhanced towards a Th1 phenotype but that the presence of Th1 CD4(+) T cells does not promote healing. This suggests that the phenotype of the CD4(+) T cells may not always be indicative of protection to L. amazonensis infection. Furthermore, our data support growing evidence that antigen-presenting cell function, such as IL-12 production, may limit the immune response in L. amazonensis-infected mice.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Células Dendríticas/inmunología , Inmunoterapia/métodos , Leishmaniasis Cutánea/terapia , Células TH1/inmunología , Animales , Células de la Médula Ósea , Enfermedad Crónica , Femenino , Interferón gamma/metabolismo , Interleucina-12/metabolismo , Subunidad p40 de la Interleucina-12 , Leishmania/inmunología , Leishmaniasis Cutánea/inmunología , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Ratones , Ratones Endogámicos C3H , Ratones SCID , Subunidades de Proteína/metabolismo , Insuficiencia del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA