Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
J Autoimmun ; 144: 103174, 2024 04.
Artículo en Inglés | MEDLINE | ID: mdl-38377868

RESUMEN

In many autoimmune diseases, autoantigen-specific Th17 cells play a pivotal role in disease pathogenesis. Th17 cells can transdifferentiate into other T cell subsets in inflammatory conditions, however, there have been no attempts to target Th17 cell plasticity using vaccines. We investigated if autoantigen-specific Th17 cells could be specifically targeted using a therapeutic vaccine approach, where antigen was formulated in all-trans retinoic acid (ATRA)-containing liposomes, permitting co-delivery of antigen and ATRA to the same target cell. Whilst ATRA was previously found to broadly reduce Th17 responses, we found that antigen formulated in ATRA-containing cationic liposomes only inhibited Th17 cells in an antigen-specific manner and not when combined with an irrelevant antigen. Furthermore, this approach shifted existing Th17 cells away from IL-17A expression and transcriptomic analysis of sorted Th17 lineage cells from IL-17 fate reporter mice revealed a shift of antigen-specific Th17 cells to exTh17 cells, expressing functional markers associated with T cell regulation and tolerance. In the experimental autoimmune encephalomyelitis (EAE) mouse model of MS, vaccination with myelin-specific (MOG) antigen in ATRA-containing liposomes reduced Th17 responses and alleviated disease. This highlights the potential of therapeutic vaccination for changing the phenotype of existing Th17 cells in the context of immune mediated diseases.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Células Th17 , Ratones , Animales , Liposomas/metabolismo , Tretinoina/farmacología , Tretinoina/metabolismo , Autoantígenos/metabolismo , Adyuvantes Inmunológicos , Inmunización , Vacunación , Fenotipo , Ratones Endogámicos C57BL , Células TH1
2.
Trends Immunol ; 37(2): 81-83, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26777728

RESUMEN

Coscolla, Copin et al. recently used comparative genomics of M. tuberculosis (Mtb) strains to show that most human T cell-recognized epitopes are hyperconserved, but bona fide variable epitopes also exist. This identification of two sets of antigens implies opposing evolutionary processes and will have an important impact on tuberculosis (TB) vaccine strategy and design.


Asunto(s)
Linfocitos T/inmunología , Tuberculosis/inmunología , Antígenos Bacterianos/inmunología , Epítopos , Humanos , Mycobacterium tuberculosis/inmunología , Vacunas contra la Tuberculosis/inmunología
3.
Trends Immunol ; 35(8): 387-95, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24875637

RESUMEN

The vaccine discovery paradigm in tuberculosis (TB) has been to mimic the natural immune response to infection. With an emphasis on interferon (IFN)-γ as the main protective cytokine, researchers have selected dominant antigens and administered them in delivery systems to promote strong T helper (Th)1 responses. However, the Bacillus Calmette-Guérin (BCG) vaccine is a strong inducer of Th1 cells, yet has limited protection in adults, and further boosting by the Modified-Vaccinia-Ankara (MVA)85A vaccine failed to enhance efficacy in a clinical trial. We review the current understanding of host-pathogen interactions in TB infection and propose that rather than boosting Th1 responses, we should focus on understanding protective immune responses that are lacking or insufficiently promoted by BCG that can intervene at critical stages of the TB life cycle.


Asunto(s)
Vacuna BCG , Interferón gamma/inmunología , Mycobacteriaceae/inmunología , Células TH1/inmunología , Vacunas contra la Tuberculosis , Tuberculosis/inmunología , Adulto , Animales , Niño , Ensayos Clínicos como Asunto , Interacciones Huésped-Patógeno , Humanos , Tuberculosis/prevención & control , Vacunas de ADN
4.
J Immunol ; 192(7): 3247-58, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24574499

RESUMEN

CD4 T cells are crucial to the control of Mycobacterium tuberculosis infection and are a key component of current vaccine strategies. Conversely, immune-mediated pathology drives disease, and recent evidence suggests that adaptive and innate responses are evolutionarily beneficial to M. tuberculosis. We compare the functionality of CD4 T cell responses mounted against dominant and cryptic epitopes of the M. tuberculosis 6-kDa early secreted Ag (ESAT-6) before and postinfection. Protective T cells against cryptic epitopes not targeted during natural infection were induced by vaccinating mice with a truncated ESAT-6 protein, lacking the dominant epitope. The ability to generate T cells that recognize multiple cryptic epitopes was MHC-haplotype dependent, including increased potential via heterologous MHC class II dimers. Before infection, cryptic epitope-specific T cells displayed enhanced proliferative capacity and delayed cytokine kinetics. After aerosol M. tuberculosis challenge, vaccine-elicited CD4 T cells expanded and recruited to the lung. In chronic infection, dominant epitope-specific T cells developed a terminal differentiated KLRG1(+)/PD-1(lo) surface phenotype that was significantly reduced in the cryptic epitope-specific T cell populations. Dominant epitope-specific T cells in vaccinated animals developed into IFN-γ- and IFN-γ,TNF-α-coproducing effector cells, characteristic of the endogenous response. In contrast, cryptic epitope-specific CD4 T cells maintained significantly greater IFN-γ(+)TNF-α(+)IL-2(+) and TNF-α(+)IL-2(+) memory-associated polyfunctionality and enhanced proliferative capacity. Vaccine-associated IL-17A production by cryptic CD4 T cells was also enhanced, but without increased neutrophilia/pathology. Direct comparison of dominant/cryptic epitope-specific CD4 T cells within covaccinated mice confirmed the superior ability of protective cryptic epitope-specific T cells to resist M. tuberculosis infection-driven T cell differentiation.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Diferenciación Celular/inmunología , Epítopos/inmunología , Mycobacterium tuberculosis/inmunología , Tuberculosis/inmunología , Animales , Antígenos Bacterianos/inmunología , Antígenos Bacterianos/metabolismo , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Proliferación Celular , Epítopos/metabolismo , Femenino , Citometría de Flujo , Interacciones Huésped-Patógeno/efectos de los fármacos , Interacciones Huésped-Patógeno/inmunología , Interferón gamma/inmunología , Interferón gamma/metabolismo , Interleucina-2/inmunología , Interleucina-2/metabolismo , Lectinas Tipo C , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/microbiología , Complejo Mayor de Histocompatibilidad/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Mycobacterium tuberculosis/metabolismo , Mycobacterium tuberculosis/fisiología , Neutrófilos/inmunología , Neutrófilos/metabolismo , Receptor de Muerte Celular Programada 1/inmunología , Receptor de Muerte Celular Programada 1/metabolismo , Receptores Inmunológicos/inmunología , Receptores Inmunológicos/metabolismo , Tuberculosis/metabolismo , Tuberculosis/microbiología , Vacunas contra la Tuberculosis/administración & dosificación , Vacunas contra la Tuberculosis/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
5.
J Immunol ; 186(3): 1627-37, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21178003

RESUMEN

Vaccines that elicit T cell responses try to mimic protective memory T cell immunity after infection by increasing the frequency of Ag-specific T cells in the immune repertoire. However, the factors that determine immunodominance during infection and after vaccination and the relation between immunodominance and protection are incompletely understood. We previously identified TB10.4(20-28) as an immunodominant epitope recognized by H2-K(d)-restricted CD8(+) T cells after M. tuberculosis infection. Here we report a second epitope, EspA(150-158), that is recognized by a substantial number of pulmonary CD8(+) T cells. The relative abundance of these T cells in the naive repertoire only partially predicts their relative frequency after M. tuberculosis infection. Furthermore, although vaccination with recombinant vaccinia virus expressing these epitopes changes their relative immunodominance in the preinfection T cell repertoire, this change is transient after challenge with M. tuberculosis. We speculate that factors intrinsic to the chronic nature of M. tuberculosis infection establishes the hierarchy of immunodominance and may explain the failure of some vaccines to provide protection.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Citotoxicidad Inmunológica , Epítopos Inmunodominantes/metabolismo , Mycobacterium tuberculosis/inmunología , Vacunas contra la Tuberculosis/administración & dosificación , Vacunas contra la Tuberculosis/inmunología , Secuencia de Aminoácidos , Animales , Presentación de Antígeno/inmunología , Antígenos Bacterianos , Proteínas Bacterianas/metabolismo , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/microbiología , Enfermedad Crónica , Femenino , Antígenos H-2/inmunología , Proteínas de Homeodominio/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Proteínas Proto-Oncogénicas/metabolismo , Factores de Transcripción/metabolismo , Tuberculosis Pulmonar/inmunología , Tuberculosis Pulmonar/prevención & control , Virus Vaccinia/inmunología
6.
NPJ Vaccines ; 8(1): 66, 2023 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-37160970

RESUMEN

The only licensed tuberculosis (TB) vaccine, Bacillus Calmette Guerin (BCG), fails to reliably protect adolescents and adults from pulmonary TB, resulting in ~1.6 million deaths annually. Protein subunit vaccines have shown promise against TB in clinical studies. Unfortunately, most subunit vaccines require multiple administrations, which increases the risk of loss to follow-up and necessitates more complex and costly logistics. Given the well-documented adjuvant effect of BCG, we hypothesized that BCG co-administration could compensate for a reduced number of subunit vaccinations. To explore this, we developed an expression-optimized version of our H107 vaccine candidate (H107e), which does not cross-react with BCG. In the CAF®01 adjuvant, a single dose of H107e induced inferior protection compared to three H107e/CAF®01 administrations. However, co-administering a single dose of H107e/CAF®01 with BCG significantly improved protection, which was equal to BCG co-administered with three H107e/CAF®01 doses. Importantly, combining BCG with a single H107e/CAF®01 dose also increased protection in previously BCG-primed animals. Overall, a single dose of H107e/CAF®01 with BCG induced long-lived immunity and triggered BCG-specific Th17 responses. These data support co-administration of BCG and subunit vaccines in both BCG naïve and BCG-primed individuals as an improved TB vaccine strategy with reduced number of vaccination visits.

7.
bioRxiv ; 2023 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-36865310

RESUMEN

After clean drinking water, vaccination is the most impactful global health intervention. However, development of new vaccines against difficult-to-target diseases is hampered by the lack of diverse adjuvants for human use. Of particular interest, none of the currently available adjuvants induce Th17 cells. Here, we develop and test an improved liposomal adjuvant, termed CAF®10b, that incorporates a TLR-9 agonist. In a head-to-head study in non-human primates (NHPs), immunization with antigen adjuvanted with CAF®10b induced significantly increased antibody and cellular immune responses compared to previous CAF® adjuvants, already in clinical trials. This was not seen in the mouse model, demonstrating that adjuvant effects can be highly species specific. Importantly, intramuscular immunization of NHPs with CAF®10b induced robust Th17 responses that were observed in circulation half a year after vaccination. Furthermore, subsequent instillation of unadjuvanted antigen into the skin and lungs of these memory animals led to significant recall responses including transient local lung inflammation observed by Positron Emission Tomography-Computed Tomography (PET-CT), elevated antibody titers, and expanded systemic and local Th1 and Th17 responses, including >20% antigen-specific T cells in the bronchoalveolar lavage. Overall, CAF®10b demonstrated an adjuvant able to drive true memory antibody, Th1 and Th17 vaccine-responses across rodent and primate species, supporting its translational potential.

8.
PLoS Pathog ; 6(6): e1000957, 2010 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-20585630

RESUMEN

Mycobacterium tuberculosis (Mtb) requires the ESX1 specialized protein secretion system for virulence, for triggering cytosolic immune surveillance pathways, and for priming an optimal CD8+ T cell response. This suggests that ESX1 might act primarily by destabilizing the phagosomal membrane that surrounds the bacterium. However, identifying the primary function of the ESX1 system has been difficult because deletion of any substrate inhibits the secretion of all known substrates, thereby abolishing all ESX1 activity. Here we demonstrate that the ESX1 substrate EspA forms a disulfide bonded homodimer after secretion. By disrupting EspA disulfide bond formation, we have dissociated virulence from other known ESX1-mediated activities. Inhibition of EspA disulfide bond formation does not inhibit ESX1 secretion, ESX1-dependent stimulation of the cytosolic pattern receptors in the infected macrophage or the ability of Mtb to prime an adaptive immune response to ESX1 substrates. However, blocking EspA disulfide bond formation severely attenuates the ability of Mtb to survive and cause disease in mice. Strikingly, we show that inhibition of EspA disulfide bond formation also significantly compromises the stability of the mycobacterial cell wall, as does deletion of the ESX1 locus or individual components of the ESX1 system. Thus, we demonstrate that EspA is a major determinant of ESX1-mediated virulence independent of its function in ESX1 secretion. We propose that ESX1 and EspA play central roles in the virulence of Mtb in vivo because they alter the integrity of the mycobacterial cell wall.


Asunto(s)
Proteínas Bacterianas/metabolismo , Pared Celular/metabolismo , Tuberculosis/patología , Virulencia , Animales , Disulfuros/metabolismo , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Mycobacterium tuberculosis/inmunología , Mycobacterium tuberculosis/metabolismo , Mycobacterium tuberculosis/patogenicidad , Fagosomas , Tasa de Supervivencia , Tuberculosis/inmunología , Tuberculosis/microbiología
9.
Nat Commun ; 12(1): 6658, 2021 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-34795205

RESUMEN

Given the encouraging clinical results of both candidate subunit vaccines and revaccination with Bacillus Calmette-Guérin (BCG) against tuberculosis (TB), there is support for combining BCG and subunit vaccination for increased efficacy. BCG and Mycobacterium tuberculosis (Mtb) share ~98% of their genome and current subunit vaccines are almost exclusively designed as BCG boosters. The goal of this study is to design a TB subunit vaccine composed of antigens not shared with BCG and explore the advantages of this design in a BCG + subunit co-administration vaccine strategy. Eight protective antigens are selected to create an Mtb-specific subunit vaccine, named H107. Whereas traditional vaccines containing BCG-shared antigens exhibit in vivo cross-reactivity to BCG, H107 shows no cross-reactivity and does not inhibit BCG colonization. Instead, co-administering H107 with BCG leads to increased adaptive responses against both H107 and BCG. Importantly, rather than expanding BCG-primed T cells, H107 broadens the overall vaccine repertoire with new T cell clones and introduces 'adjuvant-imprinted' qualities including Th17 responses and less-differentiated Th1 cells. Collectively, these features of H107 are associated with a substantial increase in long-term protection.


Asunto(s)
Mycobacterium tuberculosis/inmunología , Vacunas contra la Tuberculosis/administración & dosificación , Tuberculosis/prevención & control , Vacunación/métodos , Animales , Antígenos Bacterianos/inmunología , Vacuna BCG/administración & dosificación , Vacuna BCG/inmunología , Humanos , Inmunogenicidad Vacunal , Ratones , Mycobacterium bovis/inmunología , Células TH1/inmunología , Células Th17/inmunología , Tuberculosis/inmunología , Vacunas contra la Tuberculosis/inmunología , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/inmunología
10.
J Immunol ; 181(12): 8595-603, 2008 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-19050279

RESUMEN

Optimal immunity to Mycobacterium tuberculosis (Mtb) infection requires CD8(+) T cells, and several current Mtb vaccine candidates are being engineered to elicit enhanced CD8(+) T cell responses. However, the function of these T cells and the mechanism by which they provide protection is still unknown. We have previously shown that CD8(+) T cells specific for the mycobacterial Ags CFP10 and TB10.4 accumulate in the lungs of mice following Mtb infection and have cytolytic activity in vivo. In this study, we determine which cytolytic pathways are used by these CD8(+) T cells during Mtb infection. We find that Mtb-specific CD8(+) T cells lacking perforin have reduced cytolytic capacity in vivo. In the absence of perforin, the residual cytolytic activity is CD95 and TNFR dependent. This is particularly true in Mtb-infected lung tissue where disruption of both perforin and CD95 eliminates target cell lysis. Moreover, adoptive transfer of immune CD8(+) T cells isolated from wild-type, but not perforin-deficient mice, protect recipient mice from Mtb infection. We conclude that CD8(+) T cells elicited following Mtb infection use several cytolytic pathways in a hierarchical and compensatory manner dominated by perforin-mediated cytolysis. Finally, although several cytolytic pathways are available, adoptively transferred Mtb-specific CD8(+) T cells require perforin-mediated cytolysis to protect animals from infection. These data show that CD8(+) T cell-mediated protection during Mtb infection requires more than the secretion of IFN-gamma and specifically defines the CD8(+) cytolytic mechanisms utilized and required in vivo.


Asunto(s)
Pruebas Inmunológicas de Citotoxicidad , Epítopos de Linfocito T/inmunología , Mycobacterium tuberculosis/inmunología , Perforina/administración & dosificación , Perforina/fisiología , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/microbiología , Tuberculosis Pulmonar/prevención & control , Traslado Adoptivo , Secuencia de Aminoácidos , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/microbiología , Epítopos de Linfocito T/administración & dosificación , Femenino , Antígenos de Histocompatibilidad Clase I/administración & dosificación , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/toxicidad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Datos de Secuencia Molecular , Perforina/deficiencia , Linfocitos T Citotóxicos/trasplante , Tuberculosis Pulmonar/inmunología , Tuberculosis Pulmonar/metabolismo
11.
Mucosal Immunol ; 12(3): 816-826, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30760832

RESUMEN

T cell-mediated protection against Mycobacterium tuberculosis (Mtb) is dependent upon the ability to localize within the site of pulmonary infection and directly interact with infected cells. In turn, vaccine strategies to improve rapid T cell targeting of Mtb-infected cells after pulmonary exposure are being actively pursued. Given parenterally, the subunit vaccine H56:CAF01 elicits polyfunctional CD4 T cells that localize to the lung parenchyma and confer durable protection. Here, we find that airway mucosal boosting of parenteral H56:CAF01 immunization greatly enhances the population of long-lived lung-resident T cells (Trm) and increases early vaccine T cell responses to pulmonary Mtb challenge in multiple mouse models. However, mucosal boosting does not alter the Th1/17 vaccine signature typical of H56:CAF01 and does not further improve durable control of pulmonary infection following aerosol Mtb-challenge. Additional mucosal boosting with H56:CAF01 further enhances the Trm response without further improving protection, while blocking the recruitment of non-Trm with FTY720-treatment failed to exposed Trm-mediated protection in mucosally boosting animals. These results demonstrate the limitations of maximizing lung-localized Trm in vaccine control of pulmonary Mtb infection, especially within an immunization protocol that is already optimized for the induction of mucosal-homing Th17 cells.


Asunto(s)
Vacuna BCG/inmunología , Pulmón/inmunología , Mycobacterium tuberculosis/fisiología , Células TH1/inmunología , Células Th17/inmunología , Tuberculosis Pulmonar/inmunología , Animales , Modelos Animales de Enfermedad , Humanos , Inmunización Secundaria , Memoria Inmunológica , Pulmón/microbiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL
12.
Commun Biol ; 2: 288, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31396568

RESUMEN

Tuberculosis, caused by infection with Mycobacterium tuberculosis (Mtb), kills over 1.6 million people each year despite availability of antibiotics. The increase in drug resistant Mtb strains is a major public health emergency and host-directed therapy as adjunct to antibiotic treatment has gained increased interest. Cyclooxygenase inhibitors (COXi) are frequently used drugs to alleviate tuberculosis related symptoms. Mouse studies of acute intravenous Mtb infection have suggested a potential benefit of COXi for host-directed therapy. Here we show that COXi treatment (ibuprofen and celecoxib) is detrimental to Mtb control in different mouse models of respiratory infection. This effect links to impairments of the Type-1 helper (Th1) T-cell response as CD4 T-cells in COXi-treated animals have significantly decreased Th1 differentiation, reduced IFNγ expression and decreased protective capacity upon adoptive transfer. If confirmed in clinical trials, these findings could have major impact on global health and question the use of COXi for host-directed therapy.


Asunto(s)
Celecoxib/toxicidad , Inhibidores de la Ciclooxigenasa/toxicidad , Ibuprofeno/toxicidad , Pulmón/efectos de los fármacos , Mycobacterium tuberculosis/patogenicidad , Células TH1/efectos de los fármacos , Tuberculosis Pulmonar/microbiología , Traslado Adoptivo , Aerosoles , Animales , Carga Bacteriana , Diferenciación Celular/efectos de los fármacos , Inhibidores de la Ciclooxigenasa 2/toxicidad , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Interacciones Huésped-Patógeno , Exposición por Inhalación , Interferón gamma/inmunología , Pulmón/inmunología , Pulmón/microbiología , Activación de Linfocitos/efectos de los fármacos , Ratones Endogámicos C3H , Mycobacterium tuberculosis/inmunología , Células TH1/inmunología , Células TH1/microbiología , Células TH1/trasplante , Tuberculosis Pulmonar/inmunología
14.
Infect Immun ; 76(9): 4199-205, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18591224

RESUMEN

Mycobacterium tuberculosis infection elicits antigen-specific CD8(+) T cells that are required to control disease. It is unknown how the major histocompatibility complex class I (MHC-I) pathway samples mycobacterial antigens. CFP10 and ESAT6 are important virulence factors secreted by M. tuberculosis, and they are immunodominant targets of the human and murine T-cell response. Here, we test the hypothesis that CFP10 secretion by M. tuberculosis is required for the priming of CD8(+) T cells in vivo. Our results reveal an explicit dependence upon the bacterial secretion of the CFP10 antigen for the induction of antigen-specific CD8(+) T cells in vivo. By using well-defined M. tuberculosis mutants and carefully controlling for virulence, we show that ESX-1 function is required for the priming of CD8(+) T cells specific for CFP10. CD4(+) and CD8(+) T-cell responses to mycobacterial antigens secreted independently of ESX-1 were unaffected, suggesting that ESX-1-dependent phagosomal escape is not required for CD8(+) T-cell priming during infection. We propose that the overrepresentation of secreted proteins as dominant targets of the CD8(+) T-cell response during M. tuberculosis infection is a consequence of their preferential sampling by the MHC-I pathway. The implications of these findings should be considered in all models of antigen presentation during M. tuberculosis infection and in vaccine development.


Asunto(s)
Antígenos Bacterianos/inmunología , Proteínas Bacterianas/inmunología , Linfocitos T CD8-positivos/inmunología , Mycobacterium tuberculosis/inmunología , Animales , Antígenos Bacterianos/metabolismo , Proteínas Bacterianas/metabolismo , Femenino , Proteínas de Homeodominio/fisiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Transporte de Proteínas , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas/fisiología , Factores de Transcripción/deficiencia , Factores de Transcripción/fisiología
15.
Infect Immun ; 76(5): 2249-55, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18332205

RESUMEN

The 10-kDa culture filtrate protein (CFP-10) and 6-kDa early secretory antigen of T cells (ESAT-6) are secreted in abundance by Mycobacterium tuberculosis and are frequently recognized by T cells from infected people. The genes encoding these proteins have been deleted from the genome of the vaccine strain Mycobacterium bovis bacillus Calmette-Guérin (BCG), and it is hypothesized that these proteins are important targets of protective immunity. Indeed, vaccination with ESAT-6 elicits protective CD4+ T cells in C57BL/6 mice. We have previously shown that M. tuberculosis infection of C3H mice elicits CFP-10-specific CD8+ and CD4+ T cells. Here we demonstrate that immunization with a CFP-10 DNA vaccine stimulates a specific T-cell response only to the H-2K(k)-restricted epitope CFP-10(32-39). These CFP-10(32-39)-specific CD8+ cells undergo a rapid expansion and accumulate in the lung following challenge of immunized mice with aerosolized M. tuberculosis. Protective immunity is induced by CFP-10 DNA vaccination as measured by a CFU reduction in the lung and spleen 4 and 8 weeks after challenge with M. tuberculosis. These data demonstrate that CFP-10 is a protective antigen and that CFP-10(32-39)-specific CD8+ T cells elicited by vaccination are sufficient to mediate protection against tuberculosis.


Asunto(s)
Proteínas Bacterianas/inmunología , Linfocitos T CD8-positivos/inmunología , Infecciones por Mycobacterium/prevención & control , Mycobacterium tuberculosis/inmunología , Vacunas contra la Tuberculosis/inmunología , Vacunas de ADN/inmunología , Animales , Proteínas Bacterianas/genética , Recuento de Colonia Microbiana , Epítopos de Linfocito T/inmunología , Femenino , Interferón gamma/biosíntesis , Pulmón/inmunología , Pulmón/microbiología , Ganglios Linfáticos/inmunología , Ratones , Ratones Endogámicos C3H , Mycobacterium tuberculosis/aislamiento & purificación , Bazo/inmunología , Bazo/microbiología , Vacunas contra la Tuberculosis/genética , Vacunas de ADN/genética
16.
PLoS One ; 13(7): e0201253, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30063728

RESUMEN

There is a need for the rational design of safe and effective vaccines to protect against chronic bacterial pathogens such as Mycobacterium tuberculosis and Mycobacterium avium subsp. paratuberculosis in a number of species. One of the main challenges for vaccine development is the lack of safe adjuvants that induce protective immune responses. Cationic Adjuvant Formulation 01 (CAF01)-an adjuvant based on trehalose dibehenate (TDB) and targeting the Mincle receptor-has entered human trials based on promising pre-clinical results in a number of species. However, in cattle CAF01 only induces weak systemic immune responses. In this study, we tested the ability of three pattern recognition receptors, either alone or in combination, to activate bovine monocytes and macrophages. We found that addition of the TLR3 agonist, polyinosinic:polycytidylic acid (Poly(I:C)) to either one of the Mincle receptor agonists, TDB or monomycoloyl glycerol (MMG), enhanced monocyte activation, and calves vaccinated with CAF09 containing MMG and Poly(I:C) had increased cell-mediated and humoral immune response compared to CAF01 vaccinated animals. In contrast to the highly reactogenic Montanide ISA 61 VG, CAF09-primed T cells maintained a higher frequency of polyfunctional CD4+ T cells (IFN-γ+ TNF-α+ IL-2+). In conclusion, CAF09 supports the development of antibodies along with a high-quality cell-mediated immune response and is a promising alternative to oil-in-water adjuvant in cattle and other ruminants.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Memoria Inmunológica/efectos de los fármacos , Lectinas Tipo C/inmunología , Mycobacterium avium subsp. paratuberculosis/inmunología , Mycobacterium tuberculosis/inmunología , Linfocitos T/inmunología , Receptor Toll-Like 3/inmunología , Vacunas contra la Tuberculosis/farmacología , Adyuvantes Inmunológicos/química , Animales , Bovinos , Masculino , Paratuberculosis/inmunología , Paratuberculosis/patología , Paratuberculosis/prevención & control , Linfocitos T/patología , Vacunas contra la Tuberculosis/química , Vacunas contra la Tuberculosis/inmunología , Tuberculosis Bovina/inmunología , Tuberculosis Bovina/patología , Tuberculosis Bovina/prevención & control
17.
Front Immunol ; 9: 2717, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30534124

RESUMEN

Lipocalin-2 is a constituent of the neutrophil secondary granules and is expressed de novo by macrophages and epithelium in response to inflammation. Lipocalin-2 acts in a bacteriostatic fashion by binding iron-loaded siderophores required for bacterial growth. Mycobacterium tuberculosis (M.tb) produces siderophores that can be bound by lipocalin-2. The impact of lipocalin-2 in the innate immune response toward extracellular bacteria has been established whereas the effect on intracellular bacteria, such as M.tb, is less well-described. Here we show that lipocalin-2 surprisingly confers a growth advantage on M.tb in the early stages of infection (3 weeks post-challenge). Using mixed bone marrow chimeras, we demonstrate that lipocalin-2 derived from granulocytes, but not from epithelia and macrophages, leads to increased susceptibility to M.tb infection. In contrast, lipocalin-2 is not observed to promote mycobacterial growth at later stages of M.tb infection. We demonstrate co-localization of granulocytes and mycobacteria within the nascent granulomas at week 3 post-challenge, but not in the consolidated granulomas at week 5. We hypothesize that neutrophil-derived lipocalin-2 acts to supply a source of iron to M.tb in infected macrophages within the immature granuloma, thereby facilitating mycobacterial growth.


Asunto(s)
Granulocitos/inmunología , Granuloma/inmunología , Inmunidad Innata , Lipocalina 2/inmunología , Macrófagos/inmunología , Mycobacterium tuberculosis/inmunología , Tuberculosis/inmunología , Animales , Granulocitos/patología , Granuloma/genética , Granuloma/microbiología , Granuloma/patología , Lipocalina 2/genética , Macrófagos/microbiología , Macrófagos/patología , Ratones , Ratones Noqueados , Tuberculosis/genética , Tuberculosis/patología
18.
Crit Rev Immunol ; 26(4): 317-52, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17073557

RESUMEN

There are more cases of tuberculosis in the world today than at any other time in history. The global epidemic has generated intense interest into the immunological mechanisms that control infection. Although CD4+ T cells play a critical role in host immunity to Mycobacterium tuberculosis, there is considerable interest in understanding the role of other T cell subsets in preventing disease development following infection. CD8+ T cells are required for optimum host defense following M. tuberculosis infection, which has led to investigation into how this protective effect is mediated. A critical review of recent literature regarding the role of CD8+ T cells in protective immunity to M. tuberculosis infection is now required to address the strengths and weaknesses of these studies. In this article, we evaluate the evidence that CD8+ T cells are critical in immunity to M. tuberculosis infection. We discuss the specific mycobacterial proteins that are recognized by CD8+ T cells elicited during infection. Finally, we examine the effector mechanisms of CD8+ T cells generated during infection and synthesize recent studies to consider the protective roles that these T cells serve in vivo.


Asunto(s)
Antígenos Bacterianos/inmunología , Linfocitos T CD8-positivos/inmunología , Mycobacterium tuberculosis/inmunología , Tuberculosis/inmunología , Animales , Humanos , Ratones , Ratones Noqueados , Mycobacterium tuberculosis/metabolismo
19.
Expert Rev Vaccines ; 6(3): 441-56, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17542758

RESUMEN

Tuberculosis continues to cause considerable human morbidity and mortality worldwide, particularly in people coinfected with HIV. The emergence of multidrug resistance makes the medical treatment of tuberculosis even more difficult. Thus, the development of a tuberculosis vaccine is a global health priority. Here we review the data concerning the role of CD8+ T cells in immunity to tuberculosis and consider how CD8+ T cells can be elicited by vaccination. Many immunization strategies have the potential to elicit CD8+ T cells and we critically review the data supporting a role for vaccine-induced CD8+ T cells in protective immunity. The synergy between CD4+ and CD8+ T cells suggests that a vaccine that elicits both T-cell subsets has the best chance at preventing tuberculosis.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Vacunas contra la Tuberculosis/inmunología , Tuberculosis/prevención & control , Linfocitos T CD4-Positivos/inmunología , Infecciones por VIH/complicaciones , Humanos , Tuberculosis/inmunología , Vacunas de ADN/inmunología , Vacunas de Subunidad/inmunología
20.
J Immunol ; 177(9): 6361-9, 2006 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-17056567

RESUMEN

Whether true memory T cells develop in the face of chronic infection such as tuberculosis remains controversial. To address this question, we studied CD8+ T cells specific for the Mycobacterium tuberculosis ESAT6-related Ags TB10.3 and TB10.4. The shared epitope TB10.3/10.4(20-28) is presented by H-2 K(d), and 20-30% of the CD8+ T cells in the lungs of chronically infected mice are specific for this Ag following respiratory infection with M. tuberculosis. These TB10.3/10.4(20-28)-specific CD8+ T cells produce IFN-gamma and TNF and express CD107 on their cell surface, which indicates their likely role as CTL in vivo. Nearly all of the Ag-specific CD8+ T cells in the lungs of chronically infected mice had a T effector cell phenotype based on their low expression of CD62L and CD45RB. In contrast, a population of TB10.3/10.4(20-28)-specific CD8+ T cells was identified in the lymphoid organs that express high levels of CD62L and CD45RB. Antibiotic treatment to resolve the infection led to a contraction of the Ag-specific CD8+ T cell population and was accompanied by an increase in the proportion of CD8+ T cells with a central memory phenotype. Finally, challenge of memory-immune mice with M. tuberculosis was accompanied by significant expansion of TB10.3/10.4(20-28)-specific CD8+ T cells, which suggests that these cells are in fact functional memory T cells.


Asunto(s)
Antígenos Bacterianos/inmunología , Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica , Mycobacterium tuberculosis/inmunología , Tuberculosis Pulmonar/inmunología , Animales , Femenino , Interferón gamma/metabolismo , Selectina L/metabolismo , Antígenos Comunes de Leucocito/metabolismo , Pulmón/inmunología , Pulmón/microbiología , Tejido Linfoide/inmunología , Tejido Linfoide/microbiología , Proteína 1 de la Membrana Asociada a los Lisosomas/metabolismo , Ratones , Ratones Endogámicos BALB C , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA