Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mil Med ; 188(5-6): e1335-e1337, 2023 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-34557926

RESUMEN

We report a case of febrile Plasmodium falciparum malaria in a 36-year-old male patient occurring 14 years after immigration from and more than 12 months since a return visit to the endemic area. The critical need for awareness regarding late presentations of P. falciparum is discussed.


Asunto(s)
COVID-19 , Malaria Falciparum , Masculino , Humanos , Adulto , Plasmodium falciparum , Malaria Falciparum/complicaciones , Viaje , Emigración e Inmigración
2.
Infect Immun ; 89(11): e0016521, 2021 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-34310889

RESUMEN

Preerythrocytic vaccines prevent malaria by targeting parasites in the clinically silent sporozoite and liver stages and preventing progression to the virulent blood stages. The leading preerythrocytic vaccine, RTS,S/AS01E (Mosquirix), entered implementation programs in 2019 and targets the major sporozoite surface antigen, circumsporozoite protein (CSP). However, in phase III clinical trials, RTS,S conferred partial protection with limited durability, indicating a need to improve CSP-based vaccination. Previously, we identified highly expressed liver-stage proteins that could potentially be used in combination with CSP; they are referred to as preerythrocytic vaccine antigens (PEVAs). Here, we developed heterologous prime-boost CSP vaccination models to confer partial sterilizing immunity against Plasmodium yoelii (protein prime-adenovirus 5 [Ad5] boost) and Plasmodium berghei (DNA prime-Ad5 boost) in mice. When combined as individual antigens with P. yoelii CSP (PyCSP), three of eight P. yoelii PEVAs significantly enhanced sterile protection against sporozoite challenge, compared to PyCSP alone. Similar results were obtained when three P. berghei PEVAs and P. berghei CSP were combined in a single vaccine regimen. In general, PyCSP antibody responses were similar after CSP alone versus CSP plus PEVA vaccinations. Both P. yoelii and P. berghei CSP plus PEVA combination vaccines induced robust CD8+ T cell responses, including signature gamma interferon (IFN-γ) increases. In the P. berghei model system, IFN-γ responses were significantly higher in hepatic versus splenic CD8+ T cells. The addition of novel antigens may enhance the degree and duration of sterile protective immunity conferred by a human vaccine such as RTS,S.


Asunto(s)
Antígenos de Protozoos/inmunología , Vacunas contra la Malaria/inmunología , Proteínas Protozoarias/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Interferón gamma/biosíntesis , Activación de Linfocitos , Malaria/prevención & control , Ratones , Ratones Endogámicos BALB C , Vacunación
3.
J Clin Invest ; 131(3)2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33301427

RESUMEN

The mechanism by which only some individuals infected with Mycobacterium tuberculosis develop necrotic granulomas with progressive disease while others form controlled granulomas that contain the infection remains poorly defined. Mice carrying the sst1-suscepible (sst1S) genotype develop necrotic inflammatory lung lesions, similar to human tuberculosis (TB) granulomas, which are linked to macrophage dysfunction, while their congenic counterpart (B6) mice do not. In this study we report that (a) sst1S macrophages developed aberrant, biphasic responses to TNF characterized by superinduction of stress and type I interferon pathways after prolonged TNF stimulation; (b) the late-stage TNF response was driven via a JNK/IFN-ß/protein kinase R (PKR) circuit; and (c) induced the integrated stress response (ISR) via PKR-mediated eIF2α phosphorylation and the subsequent hyperinduction of ATF3 and ISR-target genes Chac1, Trib3, and Ddit4. The administration of ISRIB, a small-molecule inhibitor of the ISR, blocked the development of necrosis in lung granulomas of M. tuberculosis-infected sst1S mice and concomitantly reduced the bacterial burden. Hence, induction of the ISR and the locked-in state of escalating stress driven by the type I IFN pathway in sst1S macrophages play a causal role in the development of necrosis in TB granulomas. Interruption of the aberrant stress response with inhibitors such as ISRIB may offer novel host-directed therapy strategies.


Asunto(s)
Granuloma del Sistema Respiratorio/inmunología , Pulmón/inmunología , Mycobacterium tuberculosis/inmunología , Estrés Fisiológico/inmunología , Tuberculosis Pulmonar/inmunología , Animales , Modelos Animales de Enfermedad , Granuloma del Sistema Respiratorio/microbiología , Granuloma del Sistema Respiratorio/patología , Pulmón/microbiología , Pulmón/patología , Ratones , Ratones SCID , Necrosis , Tuberculosis Pulmonar/patología
4.
Front Immunol ; 9: 91, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29434602

RESUMEN

We recently identified novel Plasmodium berghei (Pb) liver stage (LS) genes that as DNA vaccines significantly reduce Pb LS parasite burden (LPB) in C57Bl/6 (B6) mice through a mechanism mediated, in part, by CD8 T cells. In this study, we sought to determine fine antigen (Ag) specificities of CD8 T cells that target LS malaria parasites. Guided by algorithms for predicting MHC class I-restricted epitopes, we ranked sequences of 32 Pb LS Ags and selected ~400 peptides restricted by mouse H-2Kb and H-2Db alleles for analysis in the high-throughput method of caged MHC class I-tetramer technology. We identified a 9-mer H-2Kb restricted CD8 T cell epitope, Kb-17, which specifically recognized and activated CD8 T cell responses in B6 mice immunized with Pb radiation-attenuated sporozoites (RAS) and challenged with infectious sporozoites (spz). The Kb-17 peptide is derived from the recently described novel protective Pb LS Ag, PBANKA_1031000 (MIF4G-like protein). Notably, immunization with the Kb-17 epitope delivered in the form of a minigene in the adenovirus serotype 5 vector reduced LPB in mice infected with spz. On the basis of our results, Kb-17 peptide was available for CD8 T cell activation and recall following immunization with Pb RAS and challenge with infectious spz. The identification of a novel MHC class I-restricted epitope from the protective Pb LS Ag, MIF4G-like protein, is crucial for advancing our understanding of immune responses to Plasmodium and by extension, toward vaccine development against malaria.


Asunto(s)
Antígenos de Protozoos/inmunología , Linfocitos T CD8-positivos/inmunología , Epítopos de Linfocito T/inmunología , Hígado/parasitología , Malaria/inmunología , Malaria/parasitología , Plasmodium berghei/inmunología , Animales , Modelos Animales de Enfermedad , Femenino , Inmunización , Interferón gamma/metabolismo , Hígado/inmunología , Malaria/metabolismo , Ratones , Plasmodium berghei/crecimiento & desarrollo
5.
PLoS One ; 11(7): e0159449, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27434123

RESUMEN

Malaria vaccine development has been hampered by the limited availability of antigens identified through conventional discovery approaches, and improvements are needed to enhance the efficacy of the leading vaccine candidate RTS,S that targets the circumsporozoite protein (CSP) of the infective sporozoite. Here we report a transcriptome-based approach to identify novel pre-erythrocytic vaccine antigens that could potentially be used in combination with CSP. We hypothesized that stage-specific upregulated genes would enrich for protective vaccine targets, and used tiling microarray to identify P. falciparum genes transcribed at higher levels during liver stage versus sporozoite or blood stages of development. We prepared DNA vaccines for 21 genes using the predicted orthologues in P. yoelii and P. berghei and tested their efficacy using different delivery methods against pre-erythrocytic malaria in rodent models. In our primary screen using P. yoelii in BALB/c mice, we found that 16 antigens significantly reduced liver stage parasite burden. In our confirmatory screen using P. berghei in C57Bl/6 mice, we confirmed 6 antigens that were protective in both models. Two antigens, when combined with CSP, provided significantly greater protection than CSP alone in both models. Based on the observations reported here, transcriptional patterns of Plasmodium genes can be useful in identifying novel pre-erythrocytic antigens that induce protective immunity alone or in combination with CSP.


Asunto(s)
Anticuerpos Antiprotozoarios/inmunología , Vacunas contra la Malaria/inmunología , Malaria Falciparum/tratamiento farmacológico , Proteínas Protozoarias/inmunología , Animales , Anticuerpos Antiprotozoarios/uso terapéutico , Antígenos de Protozoos/inmunología , Femenino , Humanos , Vacunas contra la Malaria/genética , Vacunas contra la Malaria/uso terapéutico , Malaria Falciparum/inmunología , Malaria Falciparum/parasitología , Ratones , Ratones Endogámicos C57BL , Plasmodium falciparum/inmunología , Plasmodium falciparum/patogenicidad , Plasmodium yoelii/inmunología , Vacunas de ADN/genética , Vacunas de ADN/inmunología , Vacunas de ADN/uso terapéutico
6.
Eur J Immunol ; 46(4): 885-96, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26703789

RESUMEN

MHC class I dependent CD8(+) T cells are essential for protection induced by radiation-attenuated Plasmodium sporozoites (RAS) in murine malaria models. Apart from the mechanism of activation of CD8(+) T cells specific for the circumsporozoite protein, the major sporozoite antigen (Ag), CD8(+) T cells specific for other exoerythrocytic Ags that have been shown to mediate protection have not been thoroughly investigated. Specifically, mechanisms of processing and presentation of exoerythrocytic Ags, which includes liver stage (LS) Ags, remain poorly understood. We hypothesize that as exogenous proteins, LS Ags are processed by mechanisms involving either the TAP-dependent phagosomal-to-cytosol or TAP-independent vacuolar pathway of cross-presentation. We used TAP-deficient mice to investigate whether LS Ag mediated induction of naïve CD8(+) T cells and their recall during sporozoite challenge occur by the TAP-dependent or TAP-independent pathways. On the basis of functional attributes, CD8(+) T cells were activated via the TAP-independent pathway during immunizations with Plasmodium berghei RAS; however, IFN-γ(+) CD8(+) T cells previously induced by P. berghei RAS in TAP-deficient mice failed to be recalled against sporozoite challenge and the mice became parasitemic. On the basis of these observations, we propose that TAP-associated Ag processing is indispensable for sterile protection induced with P. berghei RAS.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Presentación de Antígeno/inmunología , Antígenos de Protozoos/inmunología , Linfocitos T CD8-positivos/inmunología , Plasmodium berghei/inmunología , Esporozoítos/inmunología , Transportadoras de Casetes de Unión a ATP/inmunología , Animales , Linfocitos T CD8-positivos/citología , Diferenciación Celular/inmunología , Femenino , Antígenos de Histocompatibilidad Clase I/inmunología , Inmunización , Memoria Inmunológica/inmunología , Interferón gamma/biosíntesis , Interferón gamma/inmunología , Hígado/citología , Hígado/inmunología , Hígado/parasitología , Malaria/inmunología , Malaria/parasitología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Plasmodium berghei/efectos de la radiación , Proteínas Protozoarias/inmunología , Esporozoítos/efectos de la radiación
7.
Methods Mol Biol ; 1325: 81-9, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26450381

RESUMEN

Direct detection and quantification of liver-stage Plasmodium parasites became possible with the development of quantitative real-time PCR (qPCR). Here we describe the measurement of parasite burden in the livers of mice infected with the rodent malaria species, Plasmodium berghei and Plasmodium yoelii. This method is based on detection of expression of parasite ribosomal 18S RNA and can serve as an endpoint to accurately evaluate the efficacy of vaccines targeting the preerythrocytic stages of malaria. This approach is fast and highly reproducible and allows quantification of liver-stage parasite burden in different mouse strains and different Plasmodium species after infection with a range of sporozoite challenge doses.


Asunto(s)
Malaria/parasitología , Plasmodium berghei/aislamiento & purificación , Plasmodium yoelii/aislamiento & purificación , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Animales , Humanos , Hígado/parasitología , Hígado/patología , Malaria/genética , Malaria/patología , Ratones , Plasmodium berghei/genética , Plasmodium berghei/patogenicidad , Plasmodium yoelii/genética , Plasmodium yoelii/patogenicidad , ARN Ribosómico 18S/genética
8.
Immunol Lett ; 161(2): 189-95, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24709142

RESUMEN

Immunologic memory is one of the cardinal features of antigen-specific immune responses, and the persistence of memory cells contributes to prophylactic immunizations against infectious agents. Adequately maintained memory T and B cell pools assure a fast, effective and specific response against re-infections. However, many aspects of immunologic memory are still poorly understood, particularly immunologic memory inducible by parasites, for example, Plasmodium spp., the causative agents of malaria. For example, memory responses to Plasmodium antigens amongst residents of malaria endemic areas appear to be either inadequately developed or maintained, because persons who survive episodes of childhood malaria remain vulnerable to intermittent malaria infections. By contrast, multiple exposures of humans and laboratory rodents to radiation-attenuated Plasmodium sporozoites (γ-spz) induce sterile and long-lasting protection against experimental sporozoite challenge. Multifactorial immune mechanisms maintain this protracted and sterile protection. While the presence of memory CD4 T cell subsets has been associated with lasting protection in humans exposed to multiple bites from Anopheles mosquitoes infected with attenuated Plasmodium falciparum, memory CD8 T cells maintain protection induced with Plasmodium yoelii and Plasmodium berghei γ-spz in murine models. In this review, we discuss our observations that show memory CD8 T cells specific for antigens expressed by P. berghei liver stage parasites as an indispensable component for the maintenance of protracted protective immunity against experimental malaria infection; moreover, the provision of an Ag-depot assures a quick recall of memory T cells as IFN-γ-producing effector CD8 T cells and IL-4- producing CD4 T cells that collaborate with B cells for an effective antibody response.


Asunto(s)
Memoria Inmunológica , Malaria/inmunología , Malaria/prevención & control , Plasmodium/inmunología , Subgrupos de Linfocitos T/inmunología , Animales , Antígenos de Protozoos/inmunología , Citocinas/metabolismo , Humanos , Hígado/inmunología , Hígado/parasitología , Malaria/metabolismo , Malaria/parasitología , Parasitemia/inmunología , Parasitemia/metabolismo , Esporozoítos/inmunología , Subgrupos de Linfocitos T/metabolismo
9.
J Immunol ; 190(4): 1659-71, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23319735

RESUMEN

Mycobacterium tuberculosis is responsible for almost 2 million deaths annually. Mycobacterium bovis bacillus Calmette-Guérin, the only vaccine available against tuberculosis (TB), induces highly variable protection against TB, and better TB vaccines are urgently needed. A prerequisite for candidate vaccine Ags is that they are immunogenic and expressed by M. tuberculosis during infection of the primary target organ, that is, the lungs of susceptible individuals. In search of new TB vaccine candidate Ags, we have used a genome-wide, unbiased Ag discovery approach to investigate the in vivo expression of 2170 M. tuberculosis genes during M. tuberculosis infection in the lungs of mice. Four genetically related but distinct mouse strains were studied, representing a spectrum of TB susceptibility controlled by the supersusceptibility to TB 1 locus. We used stringent selection approaches to select in vivo-expressed M. tuberculosis (IVE-TB) genes and analyzed their expression patterns in distinct disease phenotypes such as necrosis and granuloma formation. To study the vaccine potential of these proteins, we analyzed their immunogenicity. Several M. tuberculosis proteins were recognized by immune cells from tuberculin skin test-positive, ESAT6/CFP10-responsive individuals, indicating that these Ags are presented during natural M. tuberculosis infection. Furthermore, TB patients also showed responses toward IVE-TB Ags, albeit lower than tuberculin skin test-positive, ESAT6/CFP10-responsive individuals. Finally, IVE-TB Ags induced strong IFN-γ(+)/TNF-α(+) CD8(+) and TNF-α(+)/IL-2(+) CD154(+)/CD4(+) T cell responses in PBMC from long-term latently M. tuberculosis-infected individuals. In conclusion, these IVE-TB Ags are expressed during pulmonary infection in vivo, are immunogenic, induce strong T cell responses in long-term latently M. tuberculosis-infected individuals, and may therefore represent attractive Ags for new TB vaccines.


Asunto(s)
Antígenos Bacterianos/genética , Regulación Bacteriana de la Expresión Génica/inmunología , Estudio de Asociación del Genoma Completo/métodos , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/inmunología , Subgrupos de Linfocitos T/inmunología , Tuberculosis Pulmonar/genética , Tuberculosis Pulmonar/inmunología , Animales , Antígenos Bacterianos/biosíntesis , Antígenos Bacterianos/metabolismo , Modelos Animales de Enfermedad , Marcación de Gen/métodos , Humanos , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Reproducibilidad de los Resultados , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/microbiología , Vacunas contra la Tuberculosis/genética , Vacunas contra la Tuberculosis/inmunología , Vacunas contra la Tuberculosis/uso terapéutico , Tuberculosis Pulmonar/microbiología
10.
Front Immunol ; 3: 370, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23233854

RESUMEN

Immunologic memory induced by pathogenic agents or vaccinations is inextricably linked to long-lasting protection. Adequately maintained memory T and B cell pools assure a fast, effective, and specific response against re-infections. Studies of immune responses amongst residents of malaria endemic areas suggest that memory responses to Plasmodia antigens appear to be neither adequately developed nor maintained, because persons who survive episodes of childhood malaria remain vulnerable to persistent or intermittent malaria infections. By contrast, multiple exposures of humans and laboratory rodents to radiation-attenuated Plasmodia sporozoites (γ-spz) induces sterile and long-lasting protection against experimental sporozoite challenge. Protection is associated with MHC-class I-dependent CD8 T cells, the key effectors against pre-erythrocytic stage infection. We have adopted the P. berghei γ-spz mouse model to study memory CD8 T cells that are specific for antigens expressed by Pb liver-stage (LS) parasites and are found predominantly in the liver. On the basis of phenotypic and functional characteristics, we have demonstrated that liver CD8 T cells form two subsets: CD44(hi)CD62L(lo)KLRG-1(+)CD107(+)CD127(-)CD122(lo)CD8 T effector/effector memory (T(E/EM)) cells that are the dominant IFN-γ producers and CD44(hi)CD62L(hi)KLRG-1(-)CD107(-)CD127(+)CD122(hi)CD8 T central memory (T(CM)) cells. In this review, we discuss our observations concerning the role of CD8 T(E/EM) and CD8 T(CM) cells in the maintenance of protracted protective immunity against experimental malaria infection. Finally, we present a hypothesis consistent with a model whereby intrahepatic CD8 T(CM) cells, that are maintained in part by LS-Ag depot and by IL-15-mediated survival and homeostatic proliferation, form a reservoir of cells ready for conscription to CD8 T(E/EM) cells needed to prevent re-infections.

11.
J Infect Dis ; 205(9): 1456-63, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22457289

RESUMEN

Malaria continues to be a major public health concern, and there are concerted efforts to eliminate it. The quest for a vaccine remains a top priority, and vaccines based on the circumsporozoite protein (CSP) are among the lead candidates, with the RTS,S vaccine currently undergoing phase 3 testing in Africa. Previous studies have reported anti-CSP antibody-mediated enhancement of in vitro invasion of homologous sporozoites. This effect has been shown to be concentration dependent; high-level antibodies are inhibitory, whereas low-level antibodies lead to enhancement of invasion. Nondominant shared epitopes may lead to the generation of low titers of cross-reactive antibodies that may prove to be detrimental. We report cross-species recognition of Plasmodium falciparum and Plasmodium berghei sporozoites by anti-Plasmodium vivax CSP serum samples. In addition, we report that vaccination of mice with VMP001, a P. vivax CSP vaccine candidate, reduces, not enhances, P. berghei infection in mice.


Asunto(s)
Protección Cruzada , Vacunas contra la Malaria/inmunología , Malaria/inmunología , Malaria/prevención & control , Proteínas Protozoarias/inmunología , África , Secuencia de Aminoácidos , Animales , Anticuerpos Antiprotozoarios/sangre , Epítopos/inmunología , Femenino , Inmunización , Vacunas contra la Malaria/genética , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Plasmodium berghei/inmunología , Plasmodium falciparum/inmunología , Plasmodium vivax/inmunología , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Análisis de Secuencia de ADN , Especificidad de la Especie , Esporozoítos/inmunología
12.
Am J Pathol ; 174(6): 2190-201, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19443700

RESUMEN

Significant host heterogeneity in susceptibility to tuberculosis exists both between and within mammalian species. Using a mouse model of infection with virulent Mycobacterium tuberculosis (Mtb), we identified the genetic locus sst1 that controls the progression of pulmonary tuberculosis in immunocompetent hosts. In this study, we demonstrate that within the complex, multigenic architecture of tuberculosis susceptibility, sst1 functions to control necrosis within tuberculosis lesions in the lungs; this lung-specific sst1 effect is independent of both the route of infection and genetic background of the host. Moreover, sst1-dependent necrosis was observed at low bacterial loads in the lungs during reactivation of the disease after termination of anti-tuberculosis drug therapy. We demonstrate that in sst1-susceptible hosts, nonlinked host resistance loci control both lung inflammation and production of inflammatory mediators by Mtb-infected macrophages. Although interactions of the sst1-susceptible allele with genetic modifiers determine the type of the pulmonary disease progression, other resistance loci do not abolish lung necrosis, which is, therefore, the core sst1-dependent phenotype. Sst1-susceptible mice from tuberculosis-resistant and -susceptible genetic backgrounds reproduce a clinical spectrum of pulmonary tuberculosis and may be used to more accurately predict the efficacy of anti-tuberculosis interventions in genetically heterogeneous human populations.


Asunto(s)
Predisposición Genética a la Enfermedad , Tuberculosis Pulmonar/genética , Tuberculosis Pulmonar/inmunología , Animales , Enfermedad Crónica , Modelos Animales de Enfermedad , Granuloma , Humanos , Inmunohistoquímica , Inflamación/genética , Inflamación/inmunología , Macrófagos/inmunología , Ratones , Ratones Congénicos , Ratones Endogámicos C57BL , Recurrencia , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tuberculosis Pulmonar/patología
13.
J Immunol ; 179(10): 6919-32, 2007 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-17982083

RESUMEN

Using a mouse model for genetic analysis of host resistance to virulent Mycobacterium tuberculosis, we have identified a genetic locus sst1 on mouse chromosome 1, which controls progression of pulmonary tuberculosis. In vitro, this locus had an effect on macrophage-mediated control of two intracellular bacterial pathogens, M. tuberculosis and Listeria monocytogenes. In this report, we investigated a specific function of the sst1 locus in antituberculosis immunity in vivo, especially its role in control of pulmonary tuberculosis. We found that the sst1 locus affected neither activation of Th1 cytokine-producing T lymphocytes, nor their migration to the lungs, but rather controlled an inducible NO synthase-independent mechanism of innate immunity. Although the sst1(S) macrophages responded to stimulation with IFN-gamma in vitro, their responsiveness to activation by T cells was impaired. Boosting T cell-mediated immunity by live attenuated vaccine Mycobacterium bovis bacillus Calmette-Guérin or the adoptive transfer of mycobacteria-activated CD4(+) T lymphocytes had positive systemic effect, but failed to improve control of tuberculosis infection specifically in the lungs of the sst1(S) animals. Thus, in the mouse model of tuberculosis, a common genetic mechanism of innate immunity mediated control of tuberculosis progression in the lungs and the efficiency of antituberculosis vaccine. Our data suggest that in immunocompetent humans the development of pulmonary tuberculosis and the failure of the existing vaccine to protect against it, in some cases, may be explained by a similar defect in a conserved inducible NO synthase-independent mechanism of innate immunity, either inherited or acquired.


Asunto(s)
Vacuna BCG/farmacología , Movimiento Celular/genética , Inmunidad Innata/genética , Sitios de Carácter Cuantitativo/genética , Tuberculosis Pulmonar/genética , Animales , Vacuna BCG/inmunología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/inmunología , Cromosomas/genética , Cromosomas/inmunología , Citocinas/inmunología , Modelos Animales de Enfermedad , Humanos , Inmunidad Innata/efectos de los fármacos , Listeria monocytogenes/inmunología , Listeriosis/genética , Listeriosis/inmunología , Listeriosis/patología , Ratones , Ratones Noqueados , Mycobacterium tuberculosis/inmunología , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/inmunología , Sitios de Carácter Cuantitativo/inmunología , Células TH1/inmunología , Tuberculosis Pulmonar/inmunología , Tuberculosis Pulmonar/patología , Vacunación
14.
Tuberculosis (Edinb) ; 87(2): 134-44, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17011827

RESUMEN

Dendritic cells (DC) likely play important and unique roles in the generation of protective immunity to mycobacteria. In order to clarify their contributions, bone marrow-derived DC loaded with Mycobacterium tuberculosis sonicate antigens were used to stimulate T cell proliferation both in vitro and in vivo and to vaccinate C57BL/6 mice against subsequent challenge with virulent mycobacteria. Antigen-pulsed DC developed in fetal calf serum (FCS-DC), but not DC developed in normal mouse serum (NMS-DC), stimulated significant proliferation of both naïve and immune T cells in vitro. The difference between cell populations developed in FCS and NMS in the content of CD11c(+) cells and in production of key cytokines indicated that NMS is less supportive for the development of activated DC. However, following adoptive transfer of a single dose of antigen-pulsed DC into naive recipients, NMS-DC induced T cells that proliferated in response to mycobacterial antigen, whereas FCS-DC stimulated strong non-specific proliferation. Vaccination with two doses of antigen-pulsed NMS-DC by the subcutaneous route induced significant protection against intravenous challenge with a moderate dose of virulent M. tuberculosis. DC-vaccinated mice exhibited significant reductions in bacillary loads in the lungs and spleens, and markedly reduced lung pathology. Three doses of antigen-pulsed NMS-DC induced a significant increase in survival time following high dose challenge, which correlated with a significant increase in IFN-gamma-producing cells in both lung and lymphoid tissues, as assessed by the ELISPOT assay. Taken together, these results indicate that DC play a critical role in the induction of protective resistance against virulent mycobacterial challenge accompanied by the development of antigen-reactive, IFN-gamma-producing T cells, and that their antigenic specificity is influenced by the culture conditions under which the DC are developed.


Asunto(s)
Antígenos Bacterianos/inmunología , Células Dendríticas/inmunología , Mycobacterium tuberculosis/inmunología , Vacunas contra la Tuberculosis/uso terapéutico , Tuberculosis Pulmonar/prevención & control , Traslado Adoptivo/métodos , Animales , Células de la Médula Ósea/inmunología , Antígeno CD11c/inmunología , Bovinos , División Celular/inmunología , Células Cultivadas , Recuento de Colonia Microbiana , Medios de Cultivo , Citocinas/inmunología , Modelos Animales de Enfermedad , Epítopos/inmunología , Sangre Fetal/inmunología , Pulmón/inmunología , Pulmón/microbiología , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Fenotipo , Bazo/inmunología , Bazo/microbiología , Linfocitos T/inmunología , Vacunas contra la Tuberculosis/inmunología , Tuberculosis Pulmonar/inmunología
15.
J Leukoc Biol ; 79(4): 739-46, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16415170

RESUMEN

Genetic variation in the major histocompatibility complex (MHC) influences susceptibility and immune responses to Mycobacterium tuberculosis in mice and humans, but connections among the severity of tuberculosis (TB), dynamic changes in T cell responses to mycobacteria, and MHC genetic polymorphisms are poorly characterized. The overall effect of the MHC genes on TB susceptibility and cellular responses to mycobacteria is moderate; thus, such studies provide reliable results only if congenic mouse strains bearing a variety of H2 haplotypes on an identical genetic background are analyzed. Using a panel of H2-congenic strains on the B10 background, we demonstrate that T cells from mice of three different strains, which are resistant to TB infection, readily respond by proliferation to repeated stimulations with mycobacterial sonicate, whereas T cells from three susceptible mouse strains die after the second stimulation with antigen. This difference is specific, as T cells from TB-susceptible and -resistant mouse strains do not differ in response to irrelevant antigens. The CD4/CD8 ratio in immune lymph nodes correlates strongly and inversely with TB susceptibility, being significantly lower in resistant mice as a result of an increased content of CD8+ cells. These differences between the two sets of mouse strains correlate with an elevated level of activation-induced T cell apoptosis in TB-susceptible mice and a higher proportion of activated CD44+ CD62 ligand- T cells in TB-resistant mice. These results may shed some light on the nature of the cellular basis of MHC-linked differences in susceptibility to TB.


Asunto(s)
Antígenos Bacterianos/inmunología , Apoptosis/inmunología , Antígenos H-2/inmunología , Inmunidad Innata/inmunología , Linfocitos T/inmunología , Tuberculosis Pulmonar/inmunología , Animales , Animales Congénicos/inmunología , Relación CD4-CD8 , Modelos Animales de Enfermedad , Femenino , Haplotipos , Complejo Mayor de Histocompatibilidad/genética , Complejo Mayor de Histocompatibilidad/inmunología , Ratones , Ratones Endogámicos C57BL , Mycobacterium tuberculosis/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...