Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.175
Filtrar
1.
PLoS One ; 19(9): e0307877, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39240891

RESUMEN

Tuberculosis(TB) of the Central nervous system (CNS) is a rare and highly destructive disease. The emergence of drug resistance has increased treatment difficulty, leaving the Bacillus Calmette-Guérin (BCG) vaccine as the only licensed preventative immunization available. This study focused on identifying the epitopes of PknD (Rv0931c) and Rv0986 from Mycobacterium tuberculosis(Mtb) strain H37Rv using an in silico method. The goal was to develop a therapeutic mRNA vaccine for preventing CNS TB. The vaccine was designed to be non-allergenic, non-toxic, and highly antigenic. Codon optimization was performed to ensure effective translation in the human host. Additionally, the secondary and tertiary structures of the vaccine were predicted, and molecular docking with TLR-4 was carried out. A molecular dynamics simulation confirmed the stability of the complex. The results indicate that the vaccine structure shows effectiveness. Overall, the constructed vaccine exhibits ideal physicochemical properties, immune response, and stability, laying a theoretical foundation for future laboratory experiments.


Asunto(s)
Simulación por Computador , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Mycobacterium tuberculosis , Tuberculosis del Sistema Nervioso Central , Humanos , Mycobacterium tuberculosis/inmunología , Tuberculosis del Sistema Nervioso Central/prevención & control , Tuberculosis del Sistema Nervioso Central/inmunología , Vacunas contra la Tuberculosis/inmunología , Epítopos/inmunología , Epítopos/química , Vacunas de ARNm , Vacunas Sintéticas/inmunología
2.
Indian J Tuberc ; 71(4): 453-459, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39278679

RESUMEN

The BCG vaccine, Bacille Calmette Guerin, holds the distinction of being the most widely administered vaccine. Remarkably, a century has passed since its discovery; however, puzzlingly, questions persist regarding the effectiveness of the immune response it triggers. After years of diligent observation, it has been deduced that BCG imparts immunity primarily to a specific age group, namely children. This prompts a significant query: the rationale behind BCG's limited efficacy against TB in particular age groups and populations remains elusive. Beyond vaccinations, drug therapy has emerged as an alternative route for TB prevention. Nonetheless, this approach faces challenges in the contemporary landscape, marked by the emergence of new instances of MDR-TB and XDR-TB, compounded by the financial burden of treatment. It's noteworthy that BCG remains the sole WHO-approved vaccine for TB. This comprehensive review delves into several aspects, encompassing the immune response during infection, the shortcomings of BCG in conferring immunity, and the various factors contributing to its limitations. Within this discourse, we explore potential explanations for the observed deficiencies of the BCG vaccine and consider how these insights could catalyze the development of future vaccines. The current landscape of novel vaccine development for TB is illuminated, including a spotlight on the latest vaccine candidates.


Asunto(s)
Vacuna BCG , Vacunas contra la Tuberculosis , Humanos , Vacuna BCG/inmunología , Vacunas contra la Tuberculosis/inmunología , Tuberculosis/prevención & control , Desarrollo de Vacunas , Tuberculosis Resistente a Múltiples Medicamentos/prevención & control
4.
Front Immunol ; 15: 1440935, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39108269

RESUMEN

Tuberculosis (TB) remains one of the gravest global health challenges. Mycobacterium tuberculosis (M. tuberculosis), the causative agent, employs sophisticated immune evasion and pathogenesis strategies. Its capability to thrive within immune cells and incite robust inflammatory responses prolongs infection and dissemination. Mycobacterial advanced adaptations facilitate navigation through the human immune system and present a variable antigenic profile throughout different infection stages. Investigating these strategies unfolds targeted approaches to effective vaccine development against TB. This review delves into the most advanced and exhaustive insights into the immune evasion tactics and pathogenic processes of M. tuberculosis across various infection stages. The knowledge distilled from this analysis holds the promise of guiding the creation of innovative TB vaccines and translating theoretical groundwork into practical immunological defenses.


Asunto(s)
Antígenos Bacterianos , Mycobacterium tuberculosis , Vacunas contra la Tuberculosis , Tuberculosis , Humanos , Vacunas contra la Tuberculosis/inmunología , Mycobacterium tuberculosis/inmunología , Tuberculosis/inmunología , Tuberculosis/prevención & control , Antígenos Bacterianos/inmunología , Animales , Desarrollo de Vacunas , Evasión Inmune , Interacciones Huésped-Patógeno/inmunología
5.
J Med Microbiol ; 73(8)2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39133547

RESUMEN

Introduction. Tuberculosis (TB), an infectious disease caused by Mycobacterium tuberculosis (M. tb), remains a significant global public health concern. It is crucial to develop more effective vaccines for TB in order to achieve global control of the disease. Extracellular vesicles (EVs) are spherical membrane-bound structures released by pathogens and host cells. During the course of an infection, both pathogen- and host-derived EVs are produced and play important roles in determining the course of the infection. EVs offer intriguing tools as potential vaccines due to their ability to deliver multiple pathogen or host antigens.Hypothesis /Gap Statement. We hypothesized that EVs derived from M. tb and EVs from M. tb-infected macrophages may serve as potential vaccine candidates against M. tb infection.Aim. This study aims to compare the immunogenicity and immune protection between M. tb EVs and M. tb-infected macrophage-derived EVs.Methodology. In this study, EVs were extracted from culture supernatants of M. tb and M. tb-infected macrophages, respectively. Mass spectrometry was employed to explore the antigen composition of H37Rv-Mφ-EVs and H37Rv-EVs. Cytokine profiling and antibody response assays were used to analyse the immunogenicity offered by EVs. Additionally, we used histological examination to evaluate and protective efficacy of the EVs.Results. Our results demonstrated that mice immunized by EVs released from M. tb-infected macrophages induced stronger inflammatory cytokine response than M. tb. Moreover, EVs from M. tb-infected macrophages reinforced T-cell activation and antibody response compared to M. tb EVs. Proteomic analysis revealed that EVs from M. tb-infected macrophages containing immunodominant cargos have stronger binding ability with major histocompatibility complex molecules, which may contribute to the protection from M. tb infection. Indeed, immunization of EVs released from M. tb-infected macrophages significantly reduced the bacterial load and better protection against M. tb infection than EVs from M. tb. Importantly, the selected antigens (Ag85B, ESAT-6 and the Rv0580c) from EVs of M. tb-infected macrophages exhibited effective immunogenicity.Conclusion. Our results suggested that EVs derived from M. tb-infected macrophages might serve as a proper antigenic library for vaccine candidates against M. tb challenge.


Asunto(s)
Antígenos Bacterianos , Vesículas Extracelulares , Macrófagos , Mycobacterium tuberculosis , Vacunas contra la Tuberculosis , Tuberculosis , Vesículas Extracelulares/inmunología , Mycobacterium tuberculosis/inmunología , Animales , Antígenos Bacterianos/inmunología , Vacunas contra la Tuberculosis/inmunología , Vacunas contra la Tuberculosis/administración & dosificación , Ratones , Macrófagos/inmunología , Macrófagos/microbiología , Tuberculosis/prevención & control , Tuberculosis/inmunología , Tuberculosis/microbiología , Citocinas/metabolismo , Femenino
6.
Top Antivir Med ; 32(3): 492-503, 2024 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-39142291

RESUMEN

Several novel antituberculosis agents, including long-acting injectable agents in mouse models, have shown promise in preclinical and early clinical studies. This encouraging news is offset by the failures of a tuberculosis (TB) vaccine to prevent disease recurrence and a 3-month clofazimine-based treatment regimen for drug-susceptible TB. Clinically focused insights regarding TB, mpox, and other HIV-associated infectious complications that were presented at the 2024 Conference on Retroviruses and Opportunistic Infections (CROI) are summarized in this review.


Asunto(s)
Antituberculosos , Infecciones por VIH , Tuberculosis , Humanos , Infecciones por VIH/complicaciones , Infecciones por VIH/tratamiento farmacológico , Tuberculosis/complicaciones , Tuberculosis/tratamiento farmacológico , Antituberculosos/uso terapéutico , Infecciones Oportunistas Relacionadas con el SIDA/tratamiento farmacológico , Animales , Vacunas contra la Tuberculosis
7.
PLoS Biol ; 22(8): e3002766, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39159267

RESUMEN

We report here on the characterisation in mice of a noninvasive bacille Calmette-Guérin (BCG) skin challenge model for assessing tuberculosis (TB) vaccine efficacy. Controlled human infection models (CHIMs) are valuable tools for assessing the relevant biological activity of vaccine candidates, with the potential to accelerate TB vaccine development into the clinic. TB infection poses significant constraints on the design of a CHIM using the causative agent Mycobacterium tuberculosis (Mtb). A safer alternative is a challenge model using the attenuated vaccine agent Mycobacterium bovis BCG as a surrogate for Mtb, and intradermal (skin) challenge as an alternative to pulmonary infection. We have developed a unique noninvasive imaging system based on fluorescent reporters (FluorBCG) to quantitatively measure bacterial load over time, thereby determining a relevant biological vaccine effect. We assessed the utility of this model to measure the effectiveness of 2 TB vaccines: the currently licenced BCG and a novel subunit vaccine candidate. To assess the efficacy of the skin challenge model, a nonlinear mixed-effects models was built describing the decline of fluorescence over time. The model-based analysis identified that BCG vaccination reduced the fluorescence readout of both fluorophores compared to unvaccinated mice (p < 0.001). However, vaccination with the novel subunit candidate did not alter the fluorescence decline compared to unvaccinated mice (p > 0.05). BCG-vaccinated mice that showed the reduced fluorescent readout also had a reduced bacterial burden in the lungs when challenged with Mtb. This supports the fluorescence activity in the skin as a reflection of vaccine induced functional pulmonary immune responses. This novel noninvasive approach allows for repeated measurements from the challenge site, providing a dynamic readout of vaccine induced responses over time. This BCG skin challenge model represents an important contribution to the ongoing development of controlled challenge models for TB.


Asunto(s)
Vacuna BCG , Modelos Animales de Enfermedad , Mycobacterium tuberculosis , Piel , Animales , Vacuna BCG/inmunología , Ratones , Mycobacterium tuberculosis/inmunología , Femenino , Piel/microbiología , Piel/inmunología , Tuberculosis/prevención & control , Tuberculosis/inmunología , Tuberculosis/microbiología , Eficacia de las Vacunas , Ratones Endogámicos C57BL , Carga Bacteriana , Vacunas contra la Tuberculosis/inmunología , Vacunación/métodos , Mycobacterium bovis/inmunología , Humanos
8.
EBioMedicine ; 107: 105272, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39173529

RESUMEN

BACKGROUND: MTBVAC is a live attenuated tuberculosis vaccine, currently undergoing phase III evaluation for tuberculosis prevention. In previous preclinical studies, we found that local pulmonary administration of MTBVAC via the intranasal route had a strong therapeutic effect against asthma. This effect correlated with the abrogation of allergen-specific Th2 response in the lungs. METHODS: Using different mouse models of asthma, we investigated the effect of MTBVAC administered by intravenous (IV) route and its potential as immunotherapeutic agent to induce desensitisation of allergen-specific responses at a systemic level. We explored the effects of this process in the efficacy against airway hyperresponsiveness (AHR) induced by exposure to different allergens. FINDINGS: IV MTBVAC was highly efficient at reducing AHR induced by different allergens. Additionally, IV MTBVAC was found to be well-tolerated, being progressively eliminated from the different organs analysed. From a mechanistic standpoint, we observed that MTBVAC intravenous, but not intranasal, impaired allergen-specific Th2 response in both lungs and spleen. This reduction at a systemic level correlated with long-term therapeutic protection against allergen exposure. Our results also revealed differential immunological mechanisms governing systemic and local pulmonary allergen desensitisation processes. Notably, in a cohort of patients with asthma sensitive to house dust mite (HDM), in vitro incubation of peripheral blood mononuclear cells (PBMCs) with MTBVAC prevented allergen-specific production of Th2 cytokines IL-4 and IL-5. INTERPRETATION: Altogether, our results suggest that intravenous MTBVAC could be a plausible allergen desensitising approach for treatment of asthma, and could provide long-term protection against allergen exposure. FUNDING: MCIN/AEI/10.13039/501100011033 [grants number RTI2018-097625-B-I00 and PID2022-138624OB-I00]; Consorcio Centro de Investigación Biomédica en Red- (Groups CB06/06/0020 and CB06/06/0013), Instituto de Salud Carlos III.


Asunto(s)
Administración Intravenosa , Alérgenos , Asma , Desensibilización Inmunológica , Modelos Animales de Enfermedad , Vacunas contra la Tuberculosis , Animales , Asma/inmunología , Asma/prevención & control , Ratones , Alérgenos/inmunología , Alérgenos/administración & dosificación , Humanos , Desensibilización Inmunológica/métodos , Femenino , Vacunas contra la Tuberculosis/inmunología , Vacunas contra la Tuberculosis/administración & dosificación , Pulmón/inmunología , Pulmón/patología , Células Th2/inmunología , Vacunas Atenuadas/inmunología , Vacunas Atenuadas/administración & dosificación , Citocinas/metabolismo , Ratones Endogámicos BALB C
9.
Vaccine ; 42(25): 126151, 2024 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-39089961

RESUMEN

Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB), remains one of the top three causes of death. Currently, the only licensed vaccine against TB is the bacillus Calmette-Guerin (BCG), which lacks efficacy in preventing and controlling pulmonary TB in adults. We aimed to evaluate a nasal TB vaccine formulation composed of the Mtb-specific vaccine antigen ESAT-6, an Mtb-associated protein that can trigger protective immune responses, and S100A4, a recently characterized novel mucosal adjuvant. Mice were intranasally given recombinant ESAT-6 in the presence or absence of S100A4 as an adjuvant. We have provided experimental evidence demonstrating that S100A4 admixed to ESAT-6 could induce Mtb-specific adaptive immune responses after intranasal immunization. S100A4 remarkably augmented the levels of anti-ESAT-6 IgG in serum and IgA in mucosal sites, including lung exudates, bronchoalveolar lavage fluid (BALF) and nasal lavage. Furthermore, in both lung and spleen tissues, S100A4 strongly promoted ESAT-6-specific expansion of CD4 T cells. Both CD4 and CD8 T cells from these tissues expressed increased levels of IFN-γ, TNF-α, and IL-17, cytokines critical for antimicrobial activity. Antigen-reencounter-induced T cell proliferative responses, a key vaccine performance indicator, were augmented in the spleen of S100A4-adjuvanted mice. Furthermore, CD8 T cells from the spleen and lung tissues of these mice expressed higher levels of granzyme B upon antigen re-stimulation. S100A4-adjuvanted immunization may predict good mucosal protection against TB.


Asunto(s)
Adyuvantes Inmunológicos , Administración Intranasal , Antígenos Bacterianos , Proteínas Bacterianas , Mycobacterium tuberculosis , Proteína de Unión al Calcio S100A4 , Vacunas contra la Tuberculosis , Animales , Vacunas contra la Tuberculosis/inmunología , Vacunas contra la Tuberculosis/administración & dosificación , Antígenos Bacterianos/inmunología , Proteínas Bacterianas/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Ratones , Mycobacterium tuberculosis/inmunología , Femenino , Proteína de Unión al Calcio S100A4/inmunología , Linfocitos T CD4-Positivos/inmunología , Tuberculosis/prevención & control , Tuberculosis/inmunología , Adyuvantes de Vacunas/administración & dosificación , Anticuerpos Antibacterianos/sangre , Anticuerpos Antibacterianos/inmunología , Pulmón/inmunología , Pulmón/microbiología , Linfocitos T CD8-positivos/inmunología , Inmunidad Mucosa , Inmunoglobulina A/inmunología , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Citocinas/metabolismo , Ratones Endogámicos C57BL , Bazo/inmunología , Líquido del Lavado Bronquioalveolar/inmunología , Ratones Endogámicos BALB C
10.
Eur J Pharm Biopharm ; 203: 114437, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39122053

RESUMEN

Tuberculosis (TB) has been and still is a global emergency for centuries. Prevention of disease through vaccination would have a major impact on disease prevalence, but the only available current vaccine, BCG, has insufficient impact. In this article, a novel subunit vaccine against TB was developed, using the Ag85B-ESAT6-Rv2034 fusion antigen, two adjuvants - CpG and MPLA, and a cationic pH-sensitive liposome as a delivery system, representing a new TB vaccine delivery strategy not previously reported for TB. In vitro in human dendritic cells (DCs), the adjuvanted formulation induced a significant increase in the production of (innate) cytokines and chemokines compared to the liposome without additional adjuvants. In vivo, the new vaccine administrated subcutaneously significantly reduced Mycobacterium tuberculosis (Mtb) bacterial load in the lungs and spleens of mice, significantly outperforming results from mice vaccinated with the antigen mixed with adjuvants without liposomes. In-depth analysis underpinned the vaccine's effectiveness in terms of its capacity to induce polyfunctional CD4+ and CD8+ T-cell responses, both considered essential for controlling Mtb infection. Also noteworthy was the differential abundance of various CD69+ B-cell subpopulations, which included IL17-A-producing B-cells. The vaccine stimulated robust antigen-specific antibody titers, further extending its potential as a novel protective agent against TB.


Asunto(s)
Adyuvantes Inmunológicos , Liposomas , Mycobacterium tuberculosis , Vacunas contra la Tuberculosis , Tuberculosis , Vacunas de Subunidad , Animales , Vacunas contra la Tuberculosis/administración & dosificación , Vacunas contra la Tuberculosis/inmunología , Mycobacterium tuberculosis/inmunología , Ratones , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/administración & dosificación , Humanos , Femenino , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/farmacología , Concentración de Iones de Hidrógeno , Tuberculosis/prevención & control , Tuberculosis/inmunología , Ratones Endogámicos C57BL , Células Dendríticas/inmunología , Cationes , Antígenos Bacterianos/inmunología , Antígenos Bacterianos/administración & dosificación , Citocinas/metabolismo , Linfocitos T CD8-positivos/inmunología , Pulmón/inmunología , Pulmón/microbiología , Linfocitos T CD4-Positivos/inmunología , Carga Bacteriana
11.
Microbiol Spectr ; 12(8): e0081924, 2024 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-38980025

RESUMEN

Mycobacterium kansasii is a bacterium included in non-tuberculous mycobacteria (NTM) that can cause lung disease. It shares a significant number of antigens with Mycobacterium tuberculosis (Mtb), suggesting that it has the potential to be used as a tuberculosis (TB) vaccine. Therefore, we subcutaneously vaccinated mice with reference strain, M. kansasii-ATCC12478 [M. kansasii-American Type Culture Collection (ATCC)], and clinically isolated strain, M. kansasii-SM-1 to evaluate potential as a TB vaccine by comparing with bacille Calmette-Guerin (BCG) vaccine. Ten weeks after vaccination, we evaluated immunogenicity of M. kansasii-ATCC and M. kansasii-SM-1, and M. kansasii-SM-1 immunization induces potent Mtb antigen-specific IFN-γ-producing CD4+ T cells than M. kansasii-ATCC. Upon Mtb infection, M. kansasii-SM-1 provided better protection than M. kansasii-ATCC, which was comparable to the efficacy of BCG. These results showed that the clinical strain M. kansasii-SM-1, which exhibits an enhanced Mtb antigen-specific Th1 response, shows greater vaccine efficacy compared to M. kansasii-ATCC. In this study, we demonstrated that vaccine efficacy can vary depending on the strain of M. kansasii and that its efficacy can be comparable to BCG. This suggests that M. kansasii has the potential to be a live TB vaccine candidate.IMPORTANCEMycobacterium kansasii, a non-tuberculous mycobacteria (NTM) species causing lung disease, shares key antigens with Mycobacterium tuberculosis (Mtb), indicating its potential for TB vaccine development. Subcutaneous vaccination of mice with M. kansasii strains reference strain M. kansasii-ATCC12478 [(M. kansasii-American Type Culture Collection (ATCC)] and clinically isolated strain M. kansasii-SM-1 revealed differences in immunogenicity. M. kansasii-SM-1 induced a robust Mtb antigen-specific IFN-γ-producing CD4+ T cell response compared to M. kansasii-ATCC. Additionally, M. kansasii-SM-1 conferred better protection against Mtb infection than M. kansasii-ATCC, which is comparable to bacille Calmette-Guerin (BCG). These findings underscore the variable vaccine efficacy among M. kansasii strains, with M. kansasii-SM-1 exhibiting promising potential as a live TB vaccine candidate, suggesting its comparative effectiveness to BCG.


Asunto(s)
Vacuna BCG , Mycobacterium kansasii , Mycobacterium tuberculosis , Vacunas contra la Tuberculosis , Tuberculosis , Animales , Mycobacterium kansasii/inmunología , Mycobacterium kansasii/aislamiento & purificación , Ratones , Mycobacterium tuberculosis/inmunología , Vacunas contra la Tuberculosis/inmunología , Tuberculosis/prevención & control , Tuberculosis/microbiología , Tuberculosis/inmunología , Femenino , Vacuna BCG/inmunología , Humanos , Infecciones por Mycobacterium no Tuberculosas/microbiología , Infecciones por Mycobacterium no Tuberculosas/prevención & control , Infecciones por Mycobacterium no Tuberculosas/inmunología , Linfocitos T CD4-Positivos/inmunología , Inmunogenicidad Vacunal , Antígenos Bacterianos/inmunología , Vacunación , Interferón gamma/inmunología , Interferón gamma/metabolismo , Ratones Endogámicos C57BL
12.
Immunobiology ; 229(5): 152834, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38968836

RESUMEN

Although Bacillus Calmette-Guerin (BCG) has been used in human for centuries, tuberculosis (TB) remains one of the deadliest infectious diseases.There have been remarkable successes in the field of TB vaccine research over the past decade, but the search for a better vaccine candidate is still a challenge. Extracellular vesicles (EVs) possess a multitude of properties that make them attractive candidates for the development of novel, cell-free, non-replicative, and safe vaccine system. These properties include their small size, inherent immunogenicity, ability to be taken up by immune cells, self-adjuvant capability and the comprehensive distribution of concentrated antigens. In this study, we designed a newly chimeric antigen TB vaccine (CA) with three Mycobacterium tuberculosis (M. tb) antigens that identified from extracellular vesicle derived from M. tb-infected macrophage. We confirmed that the CA stimulated a more pronounced immune response and enhanced T-cell activation, thereby providing superior protection against Mycobacterium tuberculosis infection in comparison to the bivalent antigens. Importantly, the EVs carrying CA (EVs-CA) provided enhanced protection against M. tb infection compared to unencapsulated CA antigen. Moreover, we established an EV-carried CA system (EVs-CA) and released from a transformed cell line using endogenous loading of antigen method. This method displayed the CA could efficiently package into EVs and increased concentration of this antigen. The chimeric antigen carried by EVs induced higher levels of cytokines production and specific cytotoxic T lymphocytes, resulted in enhancing antibody response and improving protective efficacy. Our findings suggested that the potential of EVs as delivery system to carry the M. tb-specific chimeric antigen for controlling Mycobacterium tuberculosis infection.


Asunto(s)
Antígenos Bacterianos , Vesículas Extracelulares , Mycobacterium tuberculosis , Vacunas contra la Tuberculosis , Tuberculosis , Vesículas Extracelulares/inmunología , Antígenos Bacterianos/inmunología , Mycobacterium tuberculosis/inmunología , Tuberculosis/inmunología , Tuberculosis/prevención & control , Animales , Humanos , Ratones , Vacunas contra la Tuberculosis/inmunología , Femenino , Macrófagos/inmunología , Activación de Linfocitos , Ratones Endogámicos C57BL , Citocinas/metabolismo
13.
Braz J Med Biol Res ; 57: e13409, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38958367

RESUMEN

Tuberculosis (TB), caused by Mycobacterium tuberculosis, remains the leading cause of mortality by a single infectious agent in the world. M. tuberculosis infection could also result in clinical chronic infection, known as latent TB infection (LTBI). Compared to the current limited treatment, several subunit vaccines showed immunotherapeutic effects and were included in clinical trials. In this study, a subunit vaccine of Ag85B with a novel mucosal adjuvant c-di-AMP (Ag85B:c-di-AMP) was delivered intranasally to a persistent M. tuberculosis H37Ra infection mouse model, which also presented the asymptomatic characteristics of LTBI. Compared with Ag85B immunization, Ag85B:c-di-AMP vaccination induced stronger humoral immune responses, significantly higher CD4+ T cells recruitment, enhanced Th1/Th2/Th17 profile response in the lung, decreased pathological lesions of the lung, and reduced M. tuberculosis load in mice. Taken together, Ag85B:c-di-AMP mucosal route immunization provided an immunotherapeutic effect on persistent M. tuberculosis H37Ra infection, and c-di-AMP, as a promising potential mucosal adjuvant, could be further used in therapeutic or prophylactic vaccine strategies for persistent M. tuberculosis infection as well as LTBI.


Asunto(s)
Adyuvantes Inmunológicos , Modelos Animales de Enfermedad , Mycobacterium tuberculosis , Vacunas contra la Tuberculosis , Animales , Adyuvantes Inmunológicos/administración & dosificación , Vacunas contra la Tuberculosis/inmunología , Vacunas contra la Tuberculosis/administración & dosificación , Mycobacterium tuberculosis/inmunología , Ratones , Femenino , Antígenos Bacterianos/inmunología , Aciltransferasas/inmunología , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/administración & dosificación , Proteínas Bacterianas/inmunología , Tuberculosis/inmunología , Tuberculosis/prevención & control , Tuberculosis Latente/inmunología , Ratones Endogámicos BALB C , Administración Intranasal
14.
Front Immunol ; 15: 1321657, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38975346

RESUMEN

Tuberculosis (TB) remains a significant global health challenge, with approximately 1.5 million deaths per year. The Bacillus Calmette-Guérin (BCG) vaccine against TB is used in infants but shows variable protection. Here, we introduce a novel approach using a double gene knockout mutant (DKO) from wild-type Mycobacterium tuberculosis (Mtb) targeting fbpA and sapM genes. DKO exhibited enhanced anti-TB gene expression in mouse antigen-presenting cells, activating autophagy and inflammasomes. This heightened immune response improved ex vivo antigen presentation to T cells. Subcutaneous vaccination with DKO led to increased protection against TB in wild-type C57Bl/6 mice, surpassing the protection observed in caspase 1/11-deficient C57Bl/6 mice and highlighting the critical role of inflammasomes in TB protection. The DKO vaccine also generated stronger and longer-lasting protection than the BCG vaccine in C57Bl/6 mice, expanding both CD62L-CCR7-CD44+/-CD127+ effector T cells and CD62L+CCR7+/-CD44+CD127+ central memory T cells. These immune responses correlated with a substantial ≥ 1.7-log10 reduction in Mtb lung burden. The DKO vaccine represents a promising new approach for TB immunization that mediates protection through autophagy and inflammasome pathways.


Asunto(s)
Macrófagos , Ratones Endogámicos C57BL , Mycobacterium tuberculosis , Vacunas contra la Tuberculosis , Tuberculosis , Animales , Mycobacterium tuberculosis/inmunología , Ratones , Macrófagos/inmunología , Tuberculosis/inmunología , Tuberculosis/prevención & control , Vacunas contra la Tuberculosis/inmunología , Antígenos Bacterianos/inmunología , Antígenos Bacterianos/genética , Inflamasomas/inmunología , Femenino , Vacuna BCG/inmunología , Autofagia/inmunología , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/genética , Modelos Animales de Enfermedad
15.
Int Immunopharmacol ; 139: 112811, 2024 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-39068754

RESUMEN

The eradication of tuberculosis remains a global challenge. Despite being the only licensed vaccine, Bacillus Calmette-Guérin (BCG) confers limited protective efficacy in adults and individuals with latent tuberculosis infections (LTBI). There is an urgent need to develop novel vaccines that can enhance the protective effect of BCG. Protein subunit vaccines have garnered significant research interest due to their safety and plasticity. Based on previous studies, we selected three antigens associated with LTBI (Rv2028c, Rv2029c, Rv3126c) and fused them with an immunodominant antigen Ag85A, resulting in the construction of a multistage protein subunit vaccine named A986. We evaluated the protective effect of recombinant protein A986 adjuvanted with MPL/QS21 as a booster vaccine for BCG against Mycobacterium tuberculosis (Mtb) infection in mice. The A986 + MPL/QS21 induced the secretion of antigen-specific Th1 (IL-2+, IFN-γ+ and TNF-α+) and Th17 (IL-17A+) cytokines in CD4+ and CD8+ T cells within the lung and spleen of mice, while also increased the frequency of central memory and effector memory T cells. Additionally, it also induced the enhanced production of IgG antibodies. Compared to BCG alone, A986 + MPL/QS21 boosting significantly augmented the proliferation of antigen-specific multifunctional T cells and effectively reduced bacterial load in infected mice. Taken together, A986 + MPL/QS21 formulation induced robust antigen-specific immune responses and provided enhanced protection against Mtb infection as a booster of BCG vaccine.


Asunto(s)
Antígenos Bacterianos , Vacuna BCG , Citocinas , Mycobacterium tuberculosis , Tuberculosis , Vacunas de Subunidad , Animales , Vacunas de Subunidad/inmunología , Mycobacterium tuberculosis/inmunología , Vacuna BCG/inmunología , Antígenos Bacterianos/inmunología , Femenino , Tuberculosis/prevención & control , Tuberculosis/inmunología , Ratones , Citocinas/metabolismo , Inmunización Secundaria , Modelos Animales de Enfermedad , Adyuvantes Inmunológicos/administración & dosificación , Vacunas contra la Tuberculosis/inmunología , Vacunas contra la Tuberculosis/administración & dosificación , Anticuerpos Antibacterianos/sangre , Anticuerpos Antibacterianos/inmunología , Ratones Endogámicos C57BL , Ratones Endogámicos BALB C , Aciltransferasas/inmunología , Pulmón/inmunología , Pulmón/microbiología , Pulmón/patología , Proteínas Bacterianas/inmunología , Humanos
16.
Arch Microbiol ; 206(8): 352, 2024 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-39012499

RESUMEN

Tuberculosis (TB) is one of the infectious diseases caused by the pathogen Mycobacterium tuberculosis that continuously threatens the global human health. Bacillus Calmette-Guérin (BCG) vaccine is the only vaccine that has been used clinically to prevent tuberculosis in recent centuries, but its limitations in preventing latent infection and reactivation of tuberculosis do not provide full protection. In this study, we selected the membrane-associated antigen Rv1513 of Mycobacterium. In order to achieve stable expression and function of the target gene, the prokaryotic expression recombinant vector pET30b-Rv1513 was constructed and expressed and purified its protein. Detection of IFN- γ levels in the peripheral blood of TB patients stimulated by whole blood interferon release assay (WBIA) and multi-microsphere flow immunofluorescence luminescence (MFCIA) revealed that the induced production of cytokines, such as IFN-γ and IL-6, was significantly higher than that in the healthy group. Rv1513 combined with adjuvant DMT (adjuvant system liposomes containing dimethyldioctadecylammonium bromide (DDA), monophospholipid A (MPL), and trehalose-660-dibenzoic acid (TDB)) was used to detect serum specific antibodies, cytokine secretion from splenic suprasplenic cell supernatants, and multifunctional T-cell levels in splenocytes in immunised mice. The levels of IFN-γ, TNF-α, and IL-2 secreted by mouse splenocytes were found in the Rv1513+DMT group and the BCG+Rv1513+DMT group. The serum levels of IgG and its subclasses and the number of IFN-γ+T cells, TNF-α+T and IFN-γ+TNF-α+T cells in the induced CD4+/CD8+T cells in mice were significantly higher than those in the BCG group, and the highest levels were found in the BCG+Rv1513+DMT group. These findings suggest that Rv1513/DMT may serve as a potential subunit vaccine candidate that may be effective as a booster vaccine after the first BCG vaccination.


Asunto(s)
Mycobacterium tuberculosis , Células TH1 , Vacunas contra la Tuberculosis , Tuberculosis , Animales , Mycobacterium tuberculosis/inmunología , Mycobacterium tuberculosis/genética , Ratones , Humanos , Células TH1/inmunología , Vacunas contra la Tuberculosis/inmunología , Vacunas contra la Tuberculosis/genética , Vacunas contra la Tuberculosis/administración & dosificación , Tuberculosis/inmunología , Tuberculosis/prevención & control , Tuberculosis/microbiología , Femenino , Antígenos Bacterianos/inmunología , Antígenos Bacterianos/genética , Citocinas/metabolismo , Citocinas/inmunología , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/genética , Interferón gamma/inmunología , Interferón gamma/metabolismo , Ratones Endogámicos BALB C , Anticuerpos Antibacterianos/sangre , Anticuerpos Antibacterianos/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Adulto
17.
Sci Rep ; 14(1): 15923, 2024 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-38987613

RESUMEN

Tuberculosis is a highly contagious disease caused by Mycobacterium tuberculosis (Mtb), which is one of the prominent reasons for the death of millions worldwide. The bacterium has a substantially higher mortality rate than other bacterial diseases, and the rapid rise of drug-resistant strains only makes the situation more concerning. Currently, the only licensed vaccine BCG (Bacillus Calmette-Guérin) is ineffective in preventing adult pulmonary tuberculosis prophylaxis and latent tuberculosis re-activation. Therefore, there is a pressing need to find novel and safe vaccines that provide robust immune defense and have various applications. Vaccines that combine epitopes from multiple candidate proteins have been shown to boost immunity against Mtb infection. This study applies an immunoinformatic strategy to generate an adequate multi-epitope immunization against Mtb employing five antigenic proteins. Potential B-cell, cytotoxic T lymphocyte, and helper T lymphocyte epitopes were speculated from the intended proteins and coupled with 50 s ribosomal L7/L12 adjuvant, and the vaccine was constructed. The vaccine's physicochemical profile demonstrates antigenic, soluble, and non-allergic. In the meantime, docking, molecular dynamics simulations, and essential dynamics analysis revealed that the multi-epitope vaccine structure interacted strongly with Toll-like receptors (TLR2 and TLR3). MM-PBSA analysis was performed to ascertain the system's intermolecular binding free energies accurately. The immune simulation was applied to the vaccine to forecast its immunogenic profile. Finally, in silico cloning was used to validate the vaccine's efficacy. The immunoinformatics analysis suggests the multi-epitope vaccine could induce specific immune responses, making it a potential candidate against Mtb. However, validation through the in-vivo study of the developed vaccine is essential to assess its efficacy and immunogenicity profile, which will assure active protection against Mtb.


Asunto(s)
Epítopos de Linfocito T , Inmunoinformática , Mycobacterium tuberculosis , Vacunas contra la Tuberculosis , Vacunas de Subunidad , Humanos , Antígenos Bacterianos/inmunología , Epítopos de Linfocito B/inmunología , Epítopos de Linfocito T/inmunología , Inmunoinformática/métodos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Mycobacterium tuberculosis/inmunología , Receptor Toll-Like 2/inmunología , Tuberculosis/prevención & control , Tuberculosis/inmunología , Vacunas contra la Tuberculosis/inmunología , Vacunas de Subunidad/inmunología
18.
Front Immunol ; 15: 1427846, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39007152

RESUMEN

To investigate how host and pathogen diversity govern immunity against Mycobacterium tuberculosis (Mtb), we performed a large-scale screen of vaccine-mediated protection against aerosol Mtb infection using three inbred mouse strains [C57BL/6 (B6), C3HeB/FeJ (C3H), Balb/c x 129/SvJ (C129F1)] and three Mtb strains (H37Rv, CDC1551, SA161) representing two lineages and distinct virulence properties. We compared three protective modalities, all of which involve inoculation with live mycobacteria: Bacillus Calmette-Guérin (BCG), the only approved TB vaccine, delivered either subcutaneously or intravenously, and concomitant Mtb infection (CoMtb), a model of pre-existing immunity in which a low-level Mtb infection is established in the cervical lymph node following intradermal inoculation. We examined lung bacterial burdens at early (Day 28) and late (Day 98) time points after aerosol Mtb challenge and histopathology at Day 98. We observed substantial heterogeneity in the reduction of bacterial load afforded by these modalities at Day 28 across the combinations and noted a strong positive correlation between bacterial burden in unvaccinated mice and the degree of protection afforded by vaccination. Although we observed variation in the degree of reduction in bacterial burdens across the nine mouse/bacterium strain combinations, virtually all protective modalities performed similarly for a given strain-strain combination. We also noted dramatic variation in histopathology changes driven by both host and bacterial genetic backgrounds. Vaccination improved pathology scores for all infections except CDC1551. However, the most dramatic impact of vaccination on lesion development occurred for the C3H-SA161 combination, where vaccination entirely abrogated the development of the large necrotic lesions that arise in unvaccinated mice. In conclusion, we find that substantial TB heterogeneity can be recapitulated by introducing variability in both host and bacterial genetics, resulting in changes in vaccine-mediated protection as measured both by bacterial burden as well as histopathology. These differences can be harnessed in future studies to identify immune correlates of vaccine efficacy.


Asunto(s)
Mycobacterium tuberculosis , Animales , Mycobacterium tuberculosis/inmunología , Mycobacterium tuberculosis/genética , Ratones , Variación Genética , Femenino , Tuberculosis/prevención & control , Tuberculosis/inmunología , Tuberculosis/microbiología , Vacunas contra la Tuberculosis/inmunología , Ratones Endogámicos C57BL , Ratones Endogámicos BALB C , Interacciones Huésped-Patógeno/inmunología , Vacuna BCG/inmunología , Pulmón/microbiología , Pulmón/patología , Pulmón/inmunología , Modelos Animales de Enfermedad , Carga Bacteriana , Vacunación
19.
Bioorg Chem ; 150: 107610, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38991488

RESUMEN

Tuberculosis (TB) continues to pose a grave threat to global health, despite relentless eradication efforts. In 1882, Robert Koch discovered that Mycobacterium tuberculosis (Mtb) is the bacterium responsible for causing tuberculosis. It is a fact that tuberculosis has claimed the lives of more than one billion people in the last few decades. It is imperative that we must take immediate and effective action to increase resources for TB research and treatment. Effective TB treatments demand an extensive investment of both time and finances, often requiring 6-9 months of rigorous antibiotic therapy. The most efficient way to control tuberculosis is by receiving a childhood Bacillus Calmette-Guérin (BCG) vaccination. Despite years of research on vaccine development, we still do not have any new approved vaccine for tuberculosis, except BCG, which is partially effective in young children. This review discusses briefly the available treatment for tuberculosis and remarkable advancements in glycoconjugate-based TB vaccine developments in recent years (2013-2024) and offers valuable direction for future research priorities.


Asunto(s)
Antituberculosos , Glicoconjugados , Mycobacterium tuberculosis , Tuberculosis , Humanos , Tuberculosis/prevención & control , Tuberculosis/tratamiento farmacológico , Glicoconjugados/química , Glicoconjugados/síntesis química , Mycobacterium tuberculosis/inmunología , Mycobacterium tuberculosis/efectos de los fármacos , Antituberculosos/química , Antituberculosos/farmacología , Antituberculosos/uso terapéutico , Vacunas contra la Tuberculosis/uso terapéutico , Desarrollo de Vacunas , Estructura Molecular , Animales
20.
Zhongguo Xue Xi Chong Bing Fang Zhi Za Zhi ; 36(2): 201-206, 2024 Mar 29.
Artículo en Chino | MEDLINE | ID: mdl-38857967

RESUMEN

Tuberculosis (TB) remains one of the biggest infectious killers worldwide. Vaccine is the most satisfactory tool for prevention of TB; however, Bacillus Calmette-Guérin (BCG), the widely used vaccine in clinical for the prevention of TB, has limitations in protective effects. Development of novel TB vaccines is therefore of urgent need. Currently, there are 15 novel TB vaccine candidates in clinical trials, including live-attenuated vaccines, inactivated vaccines, subunit vaccines and viral-vectored vaccines, which open the door for the ultimate target of the End TB Strategy. This review summarizes the latest advances in the development of TB vaccines in global clinical trials, so as to provide insights into TB control.


Asunto(s)
Ensayos Clínicos como Asunto , Vacunas contra la Tuberculosis , Tuberculosis , Humanos , Vacunas contra la Tuberculosis/inmunología , Tuberculosis/prevención & control , Desarrollo de Vacunas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA