Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
iScience ; 27(2): 108869, 2024 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-38318361

RESUMEN

Trained immunity (TI) represents a memory-like process of innate immune cells. TI can be initiated with various compounds such as fungal ß-glucan or the tuberculosis vaccine, Bacillus Calmette-Guérin. Nevertheless, considering the clinical applications of harnessing TI against infections and cancer, there is a growing need for new, simple, and easy-to-use TI inducers. Here, we demonstrate that heat-killed Mycobacterium tuberculosis (HKMtb) induces TI both in vitro and in vivo. In human monocytes, this effect represents a truly trained process, as HKMtb confers boosted inflammatory responses against various heterologous challenges, such as lipopolysaccharide (Toll-like receptor [TLR] 4 ligand) and R848 (TLR7/8 ligand). Mechanistically, HKMtb-induced TI relies on epigenetic mechanisms in a Syk/HIF-1α-dependent manner. In vivo, HKMtb induced TI when administered both systemically and intranasally, with the latter generating a more robust TI response. Summarizing, our research has demonstrated that HKMtb has the potential to act as a mucosal immunotherapy that can successfully induce trained responses.

2.
Nat Commun ; 14(1): 6090, 2023 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-37794033

RESUMEN

Intravesical administration of Bacillus Calmette-Guérin (BCG) was one of the first FDA-approved immunotherapies and remains a standard treatment for bladder cancer. Previous studies have demonstrated that intravenous (IV) administration of BCG is well-tolerated and effective in preventing tuberculosis infection in animals. Here, we examine IV BCG in several preclinical lung tumor models. Our findings demonstrate that BCG inoculation reduced tumor growth and prolonged mouse survival in models of lung melanoma metastasis and orthotopic lung adenocarcinoma. Moreover, IV BCG treatment was well-tolerated with no apparent signs of acute toxicity. Mechanistically, IV BCG induced tumor-specific CD8+ T cell responses, which were dependent on type 1 conventional dendritic cells, as well as NK cell-mediated immunity. Lastly, we also show that IV BCG has an additive effect on anti-PD-L1 checkpoint inhibitor treatment in mouse lung tumors that are otherwise resistant to anti-PD-L1 as monotherapy. Overall, our study demonstrates the potential of systemic IV BCG administration in the treatment of lung tumors, highlighting its ability to enhance immune responses and augment immune checkpoint blockade efficacy.


Asunto(s)
Neoplasias Pulmonares , Neoplasias de la Vejiga Urinaria , Ratones , Animales , Vacuna BCG , Neoplasias de la Vejiga Urinaria/patología , Linfocitos T CD8-positivos , Administración Intravenosa , Inmunidad Celular , Células Asesinas Naturales , Pulmón/patología , Neoplasias Pulmonares/tratamiento farmacológico
3.
Oncoimmunology ; 12(1): 2160094, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36567803

RESUMEN

Bacillus Calmette-Guérin (BCG), the nonpathogenic Mycobacterium bovis strain used as tuberculosis vaccine, has been successfully used as treatment for non-muscle invasive bladder cancer for decades, and suggested to potentiate cellular and humoral immune responses. However, the exact mechanism of action is not fully understood. We previously described that BCG mainly activated anti-tumor cytotoxic NK cells with upregulation of CD56 and a CD16+ phenotype. Now, we show that stimulation of human peripheral blood mononuclear cells with iBCG, a preparation based on BCG-Moreau, expands oligoclonal γδ T-cells, with a cytotoxic phenotype, together with anti-tumor CD56high CD16+ NK cells. We have used scRNA-seq, flow cytometry, and functional assays to characterize these BCG-activated γδ T-cells in detail. They had a high IFNγ secretion signature with expression of CD27+ and formed conjugates with bladder cancer cells. BCG-activated γδ T-cells proliferated strongly in response to minimal doses of cytokines and had anti-tumor functions, although not fully based on degranulation. BCG was sufficient to stimulate proliferation of γδ T-cells when cultured with other PBMC; however, BCG alone did not stimulate expansion of purified γδ T-cells. The characterization of these non-donor restricted lymphocyte populations, which can be expanded in vitro, could provide a new approach to prepare cell-based immunotherapy tools.


Asunto(s)
Antineoplásicos , Mycobacterium bovis , Neoplasias de la Vejiga Urinaria , Humanos , Vacuna BCG/uso terapéutico , Leucocitos Mononucleares , Células Asesinas Naturales , Neoplasias de la Vejiga Urinaria/terapia , Linfocitos T
4.
J Immunother Cancer ; 10(7)2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35781395

RESUMEN

BACKGROUND: Intravesical BCG is the gold-standard therapy for non-muscle invasive bladder cancer (NMIBC); however, it still fails in a significant proportion of patients, so improved treatment options are urgently needed. METHODS: Here, we compared BCG antitumoral efficacy with another live attenuated mycobacteria, MTBVAC, in an orthotopic mouse model of bladder cancer (BC). We aimed to identify both bacterial and host immunological factors to understand the antitumoral mechanisms behind effective bacterial immunotherapy for BC. RESULTS: We found that the expression of the BCG-absent proteins ESAT6/CFP10 by MTBVAC was determinant in mediating bladder colonization by the bacteria, which correlated with augmented antitumoral efficacy. We further analyzed the mechanism of action of bacterial immunotherapy and found that it critically relied on the adaptive cytotoxic response. MTBVAC enhanced both tumor antigen-specific CD4+ and CD8+ T-cell responses, in a process dependent on stimulation of type 1 conventional dendritic cells. Importantly, improved intravesical bacterial immunotherapy using MBTVAC induced eradication of fully established bladder tumors, both as a monotherapy and specially in combination with the immune checkpoint inhibitor antiprogrammed cell death ligand 1 (anti PD-L1). CONCLUSION: These results contribute to the understanding of the mechanisms behind successful bacterial immunotherapy against BC and characterize a novel therapeutic approach for BCG-unresponsive NMIBC cases.


Asunto(s)
Neoplasias de la Vejiga Urinaria , Animales , Vacuna BCG/uso terapéutico , Bacterias , Inhibidores de Puntos de Control Inmunológico , Inmunoterapia , Ratones , Neoplasias de la Vejiga Urinaria/terapia
5.
Mol Ther Nucleic Acids ; 27: 1235-1248, 2022 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-35282413

RESUMEN

Cyclic (di)nucleotides act as universal second messengers endogenously produced by several pathogens. Specifically, the roles of c-di-AMP in Mycobacterium tuberculosis immunity and virulence have been largely explored, although its contribution to the safety and efficacy of live tuberculosis vaccines is less understood. In this study, we demonstrate that the synthesis of c-di-AMP is negatively regulated by the M. tuberculosis PhoPR virulence system. Accordingly, the live attenuated tuberculosis vaccine candidate M. tuberculosis vaccine (MTBVAC), based on double phoP and fadD26 deletions, produces more than 25- and 45-fold c-di-AMP levels relative to wild-type M. tuberculosis or the current vaccine bacille Calmette-Guérin (BCG), respectively. Secretion of this second messenger was exclusively detected in MTBVAC but not in M. tuberculosis or in BCG. We also demonstrate that c-di-AMP synthesis during in vitro cultivation of M. tuberculosis is a growth-phase- and medium-dependent phenotype. To uncover the role of this metabolite in the vaccine properties of MTBVAC, we constructed and validated knockout and overproducing/oversecreting derivatives by inactivating the disA or cnpB gene, respectively. All MTBVAC derivatives elicited superior interleukin-1ß (IL-1ß) responses compared with BCG during an in vitro infection of human macrophages. However, both vaccines failed to elicit interferon ß (IFNß) activation in this cellular model. We found that increasing c-di-AMP levels remarkably correlated with a safer profile of tuberculosis vaccines in the immunodeficient mouse model. Finally, we demonstrate that overproduction of c-di-AMP due to cnpB inactivation resulted in lower protection of MTBVAC, while the absence of c-di-AMP in the MTBVAC disA derivative maintains the protective efficacy of this vaccine in mice.

6.
Sci Immunol ; 6(63): eabc2934, 2021 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-34559551

RESUMEN

Bacillus Calmette-Guerin (BCG) is an attenuated bacterial vaccine used to protect against Mycobacterium tuberculosis (Mtb) in regions where infections are highly prevalent. BCG is currently delivered by the intradermal route, but alternative routes of administration are of great interest, including intrapulmonary delivery to more closely mimic respiratory Mtb infection. In this study, mice subjected to pulmonary delivery of green fluorescent protein­tagged strains of virulent (Mtb) and attenuated (BCG) mycobacteria were studied to better characterize infected lung cell subsets. Profound differences in dissemination patterns were detected between Mtb and BCG, with a strong tendency of Mtb to disseminate from alveolar macrophages (AMs) to other myeloid subsets, mainly neutrophils and recruited macrophages. BCG mostly remained in AMs, which promoted their activation. These preactivated macrophages were highly efficient in containing Mtb bacilli upon challenge and disrupting early bacterial dissemination, which suggests a potential mechanism of protection associated with pulmonary BCG vaccination. Respiratory BCG also protected mice against a lethal Streptococcus pneumoniae challenge, suggesting that BCG-induced innate activation could confer heterologous protection against respiratory pathogens different from Mtb. BCG drove long-term activation of AMs, even after vaccine clearance, and these AMs reacted efficiently upon subsequent challenge. These results suggest the generation of a trained innate memory-like response in AMs induced by pulmonary BCG vaccination.


Asunto(s)
Vacuna BCG/inmunología , Tuberculosis Pulmonar/inmunología , Animales , Modelos Animales de Enfermedad , Pulmón/inmunología , Activación de Macrófagos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mycobacterium tuberculosis/inmunología
8.
Vaccine ; 39(50): 7277-7285, 2021 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-34238608

RESUMEN

At its 100th birthday of its first administration to a newborn, BCG has been (and continues being) an inspiration for the construction and development of hundreds of new TB vaccine candidates in the last two and a half decades. Today, 14 candidates are in clinical development inside the global TB vaccine pipeline. MTBVAC is one of these candidates. Based on a live-attenuated Mycobacterium tuberculosis clinical isolate, MTBVAC's 25 years of vaccine discovery, construction and characterisation have followed Pasteur principles, and in the process, BCG has served as a reference gold standard for establishing the safety and protective efficacy of new TB vaccine candidates. MTBVAC, which contains the antigen repertoire of M. tuberculosis, is now poised to initiate Phase 3 efficacy trials in newborns in TB-endemic countries. BCG's efficacy extends beyond that against TB, shown to confer heterologous non-specific immunity to other diseases and reduce all-cause mortality in the first months of life. Today, WHO recognises the importance that any new TB vaccine designed for administration at birth, should show similar non-specific benefits as BCG vía mechanisms of trained immunity and/or cross-reactivity of adaptive immune responses to other pathogens. Key recent studies provide strong support for MTBVAC's ability of inducing trained immunity and conferring non-specific heterologous protection similar to BCG. Research on alternative delivery routes of MTBVAC, such as a clinically feasible aerosol route, could facilitate vaccine administration for long-term TB eradication programmes in the future.


Asunto(s)
Mycobacterium tuberculosis , Vacunas contra la Tuberculosis , Tuberculosis , Vacuna BCG , Humanos , Recién Nacido , Tuberculosis/prevención & control , Vacunas Atenuadas
9.
Biology (Basel) ; 10(3)2021 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-33801808

RESUMEN

Several hundred millions of people have been diagnosed of coronavirus disease 2019 (COVID-19), causing millions of deaths and a high socioeconomic burden. SARS-CoV-2, the causative agent of COVID-19, induces both specific T- and B-cell responses, being antibodies against the virus detected a few days after infection. Passive immunization with hyperimmune plasma from convalescent patients has been proposed as a potentially useful treatment for COVID-19. Using an in-house quantitative ELISA test, we found that plasma from 177 convalescent donors contained IgG antibodies specific to the spike receptor-binding domain (RBD) of SARS-CoV-2, although at very different concentrations which correlated with previous disease severity and gender. Anti-RBD IgG plasma concentrations significantly correlated with the plasma viral neutralizing activity (VN) against SARS-CoV-2 in vitro. Similar results were found using an independent cohort of serum from 168 convalescent health workers. These results validate an in-house RBD IgG ELISA test in a large cohort of COVID-19 convalescent patients and indicate that plasma from all convalescent donors does not contain a high enough amount of anti-SARS-CoV-2-RBD neutralizing IgG to prevent SARS-CoV-2 infection in vitro. The use of quantitative anti-RBD IgG detection systems might help to predict the efficacy of the passive immunization using plasma from patients recovered from SARS-CoV-2.

10.
Front Immunol ; 12: 622995, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33708215

RESUMEN

Natural Killer cell receptors allow this heterogeneous immune population to efficiently fight both tumors and infection, so their use as immunotherapy agents is an active field of research. Cytokine activation, particularly by myeloid cell-derived IL15, can induce potent NK anti-tumor responses. While studying the mechanism of action of intravesical instillations of Bacille Calmette-Guérin (BCG) as therapy for patients with high risk non-muscle invasive bladder cancer, we showed that BCG can activate a cytotoxic CD56bright NK cell population which efficiently recognized bladder cancer cells. This pioneer immunotherapy provides an invaluable model to understand the role of different immune populations in tumor elimination. However, during the propagation of BCG worldwide a large number of genetically diverse BCG substrains developed. Here, we investigated the capacity of different BCG substrains to promote NK cell activation and confirmed that they were able to activate lymphocytes. Tice, Connaught and Moreau were the substrains with a stronger NK activation effect as measured by CD56 upregulation. Surprisingly, dead mycobacteria also stimulated PBMC cultures and we further demonstrate here that subcellular fractions of BCG-Tice, in the absence of live mycobacteria, could also induce an NK cell response. Lipids from BCG-Tice, but not from Mycobacterium bovis, stimulated NK cell activation and degranulation, whereas the aqueous fraction of either bacteria did not activate lymphocytes. However, delipidated BCG-Tice bacteria were able to activate effector cells (CD3+CD56+ and NK, CD3-CD56+). These data demonstrate that different components of mycobacteria can stimulate different immune subpopulations resulting in phenotypes suitable for cancer elimination.


Asunto(s)
Antineoplásicos/inmunología , Vacuna BCG/inmunología , Degranulación de la Célula , Inmunoterapia , Células Asesinas Naturales/inmunología , Activación de Linfocitos , Mycobacterium bovis/inmunología , Neoplasias de la Vejiga Urinaria/terapia , Vacuna BCG/genética , Complejo CD3/metabolismo , Antígeno CD56/metabolismo , Proliferación Celular , Técnicas de Cocultivo , Humanos , Células K562 , Células Asesinas Naturales/metabolismo , Mycobacterium bovis/genética , Microambiente Tumoral , Neoplasias de la Vejiga Urinaria/inmunología , Neoplasias de la Vejiga Urinaria/metabolismo , Vacunas de Productos Inactivados/genética , Vacunas de Productos Inactivados/inmunología
11.
EBioMedicine ; 65: 103254, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33711798

RESUMEN

BACKGROUND: The Bacillus Calmette-Guérin (BCG), the only vaccine against tuberculosis (TB) currently in use, has shown beneficial effects against unrelated infections and to enhance immune responses to vaccines. However, there is little evidence regarding the influence of BCG vaccination on pertussis. METHODS: Here, we studied the ability of BCG to improve the immune responses to diphtheria, tetanus, and acellular (DTaP) or whole-cell pertussis (DTwP) vaccination in a mouse model. We included MTBVAC, an experimental live-attenuated vaccine derived from Mycobacterium tuberculosis, in our studies to explore if it presents similar heterologous immunity as BCG. Furthermore, we explored the potential effect of routine BCG vaccination on pertussis incidence worldwide. FINDINGS: We found that both BCG and MTBVAC when administered before DTaP, triggered Th1 immune responses against diphtheria, tetanus, and pertussis in mice. Immunization with DTaP alone failed to trigger a Th1 response, as measured by the production of IFN-γ. Humoral responses against DTaP antigens were also enhanced by previous immunization with BCG or MTBVAC. Furthermore, exploration of human epidemiological data showed that pertussis incidence was 10-fold lower in countries that use DTaP and BCG compared to countries that use only DTaP. INTERPRETATION: BCG vaccination may have a beneficial impact on the protection against pertussis conferred by DTaP. Further randomized controlled trials are needed to properly define the impact of BCG on pertussis incidence in a controlled setting. This could be a major finding that would support changes in immunization policies. FUNDING: This work was supported by the Ministry of "Economía y Competitividad"; European Commission H2020 program, "Gobierno de Aragón"; CIBERES; "Fundação Butantan"; Instituto de Salud Carlos III and "Fondo FEDER".


Asunto(s)
Vacuna BCG/administración & dosificación , Vacunas contra Difteria, Tétanos y Tos Ferina Acelular/administración & dosificación , Inmunidad Humoral , Tos Ferina/prevención & control , Animales , Anticuerpos Antibacterianos/sangre , Anticuerpos Antibacterianos/inmunología , Antígenos Bacterianos/inmunología , Difteria/inmunología , Difteria/prevención & control , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Incidencia , Interferón gamma/metabolismo , Ratones , Ratones Endogámicos BALB C , Tétanos/inmunología , Tétanos/prevención & control , Células TH1/citología , Células TH1/inmunología , Células TH1/metabolismo , Vacunación , Tos Ferina/epidemiología , Tos Ferina/inmunología
12.
Cell Rep Med ; 2(1): 100185, 2021 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-33521699

RESUMEN

BCG vaccination can strengthen protection against pathogens through the induction of epigenetic and metabolic reprogramming of innate immune cells, a process called trained immunity. We and others recently demonstrated that mucosal or intravenous BCG better protects rhesus macaques from Mycobacterium tuberculosis infection and TB disease than standard intradermal vaccination, correlating with local adaptive immune signatures. In line with prior mouse data, here, we show in rhesus macaques that intravenous BCG enhances innate cytokine production associated with changes in H3K27 acetylation typical of trained immunity. Alternative delivery of BCG does not alter the cytokine production of unfractionated bronchial lavage cells. However, mucosal but not intradermal vaccination, either with BCG or the M. tuberculosis-derived candidate MTBVAC, enhances innate cytokine production by blood- and bone marrow-derived monocytes associated with metabolic rewiring, typical of trained immunity. These results provide support to strategies for improving TB vaccination and, more broadly, modulating innate immunity via mucosal surfaces.


Asunto(s)
Vacuna BCG/administración & dosificación , Inmunidad Mucosa , Mycobacterium tuberculosis/inmunología , Mucosa Respiratoria/inmunología , Vacunas contra la Tuberculosis/administración & dosificación , Tuberculosis Pulmonar/prevención & control , Acetilación , Administración Intranasal , Animales , Médula Ósea/efectos de los fármacos , Médula Ósea/inmunología , Médula Ósea/microbiología , Reprogramación Celular/genética , Reprogramación Celular/inmunología , Femenino , Regulación de la Expresión Génica , Histonas/genética , Histonas/inmunología , Inyecciones Intravenosas , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/microbiología , Macaca mulatta , Masculino , Monocitos/efectos de los fármacos , Monocitos/inmunología , Monocitos/microbiología , Mycobacterium tuberculosis/patogenicidad , Mucosa Respiratoria/microbiología , Tuberculosis Pulmonar/genética , Tuberculosis Pulmonar/inmunología , Tuberculosis Pulmonar/microbiología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
13.
Cell Rep Med ; 2(1): 100187, 2021 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-33521701

RESUMEN

To fight tuberculosis, better vaccination strategies are needed. Live attenuated Mycobacterium tuberculosis-derived vaccine, MTBVAC, is a promising candidate in the pipeline, proven to be safe and immunogenic in humans so far. Independent studies have shown that pulmonary mucosal delivery of Bacillus Calmette-Guérin (BCG), the only tuberculosis (TB) vaccine available today, confers superior protection over standard intradermal immunization. Here we demonstrate that mucosal MTBVAC is well tolerated, eliciting polyfunctional T helper type 17 cells, interleukin-10, and immunoglobulins in the airway and yielding a broader antigenic profile than BCG in rhesus macaques. Beyond our previous work, we show that local immunoglobulins, induced by MTBVAC and BCG, bind to M. tuberculosis and enhance pathogen uptake. Furthermore, after pulmonary vaccination, but not M. tuberculosis infection, local T cells expressed high levels of mucosal homing and tissue residency markers. Our data show that pulmonary MTBVAC administration has the potential to enhance its efficacy and justifies further exploration of mucosal vaccination strategies in preclinical efficacy studies.


Asunto(s)
Vacuna BCG/administración & dosificación , Inmunidad Mucosa , Mycobacterium tuberculosis/inmunología , Mucosa Respiratoria/inmunología , Vacunas contra la Tuberculosis/administración & dosificación , Tuberculosis Pulmonar/prevención & control , Administración Intranasal , Animales , Reprogramación Celular/genética , Reprogramación Celular/inmunología , Femenino , Regulación de la Expresión Génica , Inyecciones Intradérmicas , Interleucina-10/genética , Interleucina-10/inmunología , Interleucina-17/genética , Interleucina-17/inmunología , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/microbiología , Macaca mulatta , Masculino , Monocitos/efectos de los fármacos , Monocitos/inmunología , Monocitos/microbiología , Mycobacterium tuberculosis/patogenicidad , Mucosa Respiratoria/microbiología , Células TH1/inmunología , Células TH1/microbiología , Células Th17/inmunología , Células Th17/microbiología , Tuberculosis Pulmonar/genética , Tuberculosis Pulmonar/inmunología , Tuberculosis Pulmonar/microbiología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
14.
EBioMedicine ; 64: 103186, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33478923

RESUMEN

BACKGROUND: Substantial recent advances in the comprehension of the molecular and cellular mechanisms behind asthma have evidenced the importance of the lung immune environment for disease outcome, making modulation of local immune responses an attractive therapeutic target against this pathology. Live attenuated mycobacteria, such as the tuberculosis vaccine BCG, have been classically linked with a type 1 response, and proposed as possible modulators of the type 2 response usually associated with asthma. METHODS: In this study we used different acute and chronic murine models of asthma to investigate the therapeutic efficacy of intranasal delivery of the live tuberculosis vaccines BCG and MTBVAC by regulating the lung immune environment associated with airway hyperresponsiveness (AHR). FINDINGS: Intranasal administration of BCG, or the novel tuberculosis vaccine candidate MTBVAC, abrogated AHR-associated hallmarks, including eosinophilia and lung remodeling. This correlated with the re-polarization of allergen-induced M2 macrophages towards an M1 phenotype, as well as with the induction of a strong allergen-specific Th1 response. Importantly, vaccine treatment was effective in a scenario of established chronic asthma where a strong eosinophil infiltration was already present prior to immunization. We finally compared the nebulization efficiency of clinical formulations of MTBVAC and BCG using a standard commercial nebulizer for potential aerosol application. INTERPRETATION: Our results demonstrate that pulmonary live tuberculosis vaccines efficiently revert established asthma in mice. These data support the further exploration of this approach as potential therapy against asthma. FUNDING: Spanish Ministry of Science [grant numbers: BIO2014-5258P, RTI2018-097625-B-I00], Instituto de Salud Carlos III, Gobierno de Aragón/Fondo Social Europeo, University of Zaragoza [grant number: JIUZ-2018-BIO-01].


Asunto(s)
Asma/inmunología , Asma/terapia , Vacunas contra la Tuberculosis/uso terapéutico , Vacunas Atenuadas/uso terapéutico , Administración Intranasal , Remodelación de las Vías Aéreas (Respiratorias)/inmunología , Alérgenos/inmunología , Animales , Vacuna BCG , Biomarcadores , Microambiente Celular/inmunología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Eosinófilos/inmunología , Eosinófilos/metabolismo , Eosinófilos/patología , Femenino , Inmunización , Ratones , Ovalbúmina/inmunología , Vacunas contra la Tuberculosis/administración & dosificación , Vacunas Atenuadas/administración & dosificación
15.
PLoS Pathog ; 16(12): e1009061, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33347499

RESUMEN

Species belonging to the Mycobacterium tuberculosis Complex (MTBC) show more than 99% genetic identity but exhibit distinct host preference and virulence. The molecular genetic changes that underly host specificity and infection phenotype within MTBC members have not been fully elucidated. Here, we analysed RD900 genomic region across MTBC members using whole genome sequences from 60 different MTBC strains so as to determine its role in the context of MTBC evolutionary history. The RD900 region comprises two homologous genes, pknH1 and pknH2, encoding a serine/threonine protein kinase PknH flanking the tbd2 gene. Our analysis revealed that RD900 has been independently lost in different MTBC lineages and different strains, resulting in the generation of a single pknH gene. Importantly, all the analysed M. bovis and M. caprae strains carry a conserved deletion within a proline rich-region of pknH, independent of the presence or absence of RD900. We hypothesized that deletion of pknH proline rich-region in M. bovis may affect PknH function, having a potential role in its virulence and evolutionary adaptation. To explore this hypothesis, we constructed two M. bovis 'knock-in' strains containing the M. tuberculosis pknH gene. Evaluation of their virulence phenotype in mice revealed a reduced virulence of both M. bovis knock-in strains compared to the wild type, suggesting that PknH plays an important role in the differential virulence phenotype of M. bovis vs M. tuberculosis.


Asunto(s)
Proteínas Bacterianas/genética , Interacciones Microbiota-Huesped/genética , Mycobacterium tuberculosis/genética , Proteínas Serina-Treonina Quinasas/genética , Animales , Proteínas Bacterianas/metabolismo , Femenino , Genómica , Masculino , Ratones , Ratones Endogámicos C57BL , Mycobacterium tuberculosis/patogenicidad , Polimorfismo Genético/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Virulencia/genética
16.
Front Microbiol ; 11: 1339, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32625195

RESUMEN

Vaccination through the natural route of infection represents an attractive immunization strategy in vaccinology. In the case of tuberculosis, vaccine delivery by the respiratory route has regained interest in recent years, showing efficacy in different animal models. In this context, respiratory vaccination triggers lung immunological mechanisms which are omitted when vaccines are administered by parenteral route. However, contribution of mucosal antibodies to vaccine- induced protection has been poorly studied. In the present study, we evaluated in mice and non-human primates (NHP) a novel whole cell inactivated vaccine (MTBVAC HK), by mucosal administration. MTBVAC HK given by intranasal route to BCG-primed mice substantially improved the protective efficacy conferred by subcutaneous BCG only. Interestingly, this improved protection was absent in mice lacking polymeric Ig receptor (pIgR), suggesting a crucial role of mucosal secretory immunoglobulins in protective immunity. Our study in NHP confirmed the ability of MTBVAC HK to trigger mucosal immunoglobulins. Importantly, in vitro assays demonstrated the functionality of these immunoglobulins to induce M. tuberculosis opsonization in the presence of human macrophages. Altogether, our results suggest that mucosal immunoglobulins can be induced by vaccination to improve protection against tuberculosis and therefore, they represent a promising target for next generation tuberculosis vaccines.

17.
EBioMedicine ; 55: 102761, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32361249

RESUMEN

BACKGROUND: Human tuberculosis (TB) is caused by a plethora of Mycobacterium tuberculosis complex (MTBC) strains belonging to seven phylogenetic branches. Lineages 2, 3 and 4 are considered "modern" branches of the MTBC responsible for the majority of worldwide TB. Since the current BCG vaccine confers variable protection against pulmonary TB, new candidates are investigated. MTBVAC is the unique live attenuated vaccine based on M. tuberculosis in human clinical trials. METHODS: MTBVAC was originally constructed by unmarked phoP and fadD26 deletions in a clinical isolate belonging to L4. Here we construct new vaccines based on isogenic gene deletions in clinical isolates of the L2 and L3 modern lineages. These three vaccine candidates were characterized at molecular level and also in animal experiments of protection and safety. FINDINGS: Safety studies in immunocompromised mice showed that MTBVAC-L2 was less attenuated than BCG Pasteur, while the original MTBVAC was found even more attenuated than BCG and MTBVAC-L3 showed an intermediate phenotype. The three MTBVAC candidates showed similar or superior protection compared to BCG in immunocompetent mice vaccinated with each MTBVAC candidate and challenged with three representative strains of the modern lineages. INTERPRETATION: MTBVAC vaccines, based on double phoP and fadD26 deletions, protect against TB independently of the phylogenetic linage used as template strain for their construction. Nevertheless, lineage L4 confers the best safety profile. FUNDING: European Commission (TBVAC2020, H2020-PHC-643381), Spanish Ministry of Science (RTI2018-097625-B-I00), Instituto de Salud Carlos III (PI18/0336), Gobierno de Aragón/Fondo Social Europeo and the French National Research Council (ANR-10-LABX-62-IBEID, ANR-16-CE35-0009, ANR-16-CE15-0003).


Asunto(s)
Proteínas Bacterianas/inmunología , Ligasas/inmunología , Mycobacterium tuberculosis/inmunología , Vacunas contra la Tuberculosis/administración & dosificación , Tuberculosis Pulmonar/genética , Tuberculosis Pulmonar/prevención & control , Animales , Vacuna BCG/administración & dosificación , Vacuna BCG/biosíntesis , Vacuna BCG/genética , Proteínas Bacterianas/genética , Femenino , Eliminación de Gen , Expresión Génica , Antecedentes Genéticos , Humanos , Inmunogenicidad Vacunal , Ligasas/deficiencia , Ligasas/genética , Ratones , Ratones SCID , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/patogenicidad , Seguridad del Paciente , Análisis de Supervivencia , Vacunas contra la Tuberculosis/biosíntesis , Vacunas contra la Tuberculosis/genética , Tuberculosis Pulmonar/inmunología , Tuberculosis Pulmonar/mortalidad , Vacunación , Vacunas Atenuadas , Población Blanca
18.
J Immunother Cancer ; 8(1)2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32241808

RESUMEN

BACKGROUND: Elimination of cancer cells by some stimuli like chemotherapy and radiotherapy activates anticancer immunity after the generation of damage-associated molecular patterns, a process recently named immunogenic cell death (ICD). Despite the recent advances in cancer immunotherapy, very little is known about the immunological consequences of cell death activated by cytotoxic CD8+ T (Tc) cells on cancer cells, that is, if Tc cells induce ICD on cancer cells and the molecular mechanisms involved. METHODS: ICD induced by Tc cells on EL4 cells was analyzed in tumor by vaccinating mice with EL4 cells killed in vitro or in vivo by Ag-specific Tc cells. EL4 cells and mutants thereof overexpressing Bcl-XL or a dominant negative mutant of caspase-3 and wild-type mice, as well as mice depleted of Tc cells and mice deficient in perforin, TLR4 and BATF3 were used. Ex vivo cytotoxicity of spleen cells from immunized mice was analyzed by flow cytometry. Expression of ICD signals (calreticulin, HMGB1 and interleukin (IL)-1ß) was analyzed by flow cytometry and ELISA. RESULTS: Mice immunized with EL4.gp33 cells killed in vitro or in vivo by gp33-specific Tc cells were protected from parental EL4 tumor development. This result was confirmed in vivo by using ovalbumin (OVA) as another surrogate antigen. Perforin and TLR4 and BATF3-dependent type 1 conventional dendritic cells (cDC1s) were required for protection against tumor development, indicating cross-priming of Tc cells against endogenous EL4 tumor antigens. Tc cells induced ICD signals in EL4 cells. Notably, ICD of EL4 cells was dependent on caspase-3 activity, with reduced antitumor immunity generated by caspase-3-deficient EL4 cells. In contrast, overexpression of Bcl-XL in EL4 cells had no effect on induction of Tc cell antitumor response and protection. CONCLUSIONS: Elimination of tumor cells by Ag-specific Tc cells is immunogenic and protects against tumor development by generating new Tc cells against EL4 endogenous antigens. This finding helps to explain the enhanced efficacy of T cell-dependent immunotherapy and provide a molecular basis to explain the epitope spread phenomenon observed during vaccination and chimeric antigen receptor (CAR)-T cell therapy. In addition, they suggest that caspase-3 activity in the tumor may be used as a biomarker to predict cancer recurrence during T cell-dependent immunotherapies.


Asunto(s)
Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Caspasa 3/metabolismo , Células Dendríticas/inmunología , Linfoma de Células T/inmunología , Linfoma de Células T/patología , Linfocitos T Citotóxicos/inmunología , Animales , Linfocitos T CD8-positivos/metabolismo , Muerte Celular , Células Dendríticas/metabolismo , Linfoma de Células T/metabolismo , Ratones , Ratones Endogámicos C57BL , Linfocitos T Citotóxicos/metabolismo
19.
PLoS Pathog ; 16(4): e1008404, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32240273

RESUMEN

Among infectious diseases, tuberculosis is the leading cause of death worldwide, and represents a serious threat, especially in developing countries. The protective effects of Bacillus Calmette-Guerin (BCG), the current vaccine against tuberculosis, have been related not only to specific induction of T-cell immunity, but also with the long-term epigenetic and metabolic reprogramming of the cells from the innate immune system through a process termed trained immunity. Here we show that MTBVAC, a live attenuated strain of Mycobacterium tuberculosis, safe and immunogenic against tuberculosis antigens in adults and newborns, is also able to generate trained immunity through the induction of glycolysis and glutaminolysis and the accumulation of histone methylation marks at the promoters of proinflammatory genes, facilitating an enhanced response after secondary challenge with non-related bacterial stimuli. Importantly, these findings in human primary myeloid cells are complemented by a strong MTBVAC-induced heterologous protection against a lethal challenge with Streptococcus pneumoniae in an experimental murine model of pneumonia.


Asunto(s)
Modelos Animales de Enfermedad , Inmunidad Innata/inmunología , Monocitos/inmunología , Mycobacterium tuberculosis/inmunología , Neumonía/prevención & control , Vacunas contra la Tuberculosis/administración & dosificación , Tuberculosis/prevención & control , Animales , Vacuna BCG/administración & dosificación , Vacuna BCG/inmunología , Células Cultivadas , Reprogramación Celular , Femenino , Humanos , Inmunidad Innata/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Mycobacterium tuberculosis/efectos de los fármacos , Neumonía/inmunología , Neumonía/microbiología , Tuberculosis/inmunología , Tuberculosis/microbiología , Vacunación
20.
Vet Res ; 50(1): 82, 2019 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-31615555

RESUMEN

Effective vaccines against tuberculosis (TB) are needed in order to prevent TB transmission in human and animal populations. Evaluation of TB vaccines may be facilitated by using reliable animal models that mimic host pathophysiology and natural transmission of the disease as closely as possible. In this study, we evaluated the immunogenicity and efficacy of two attenuated vaccines, BCG and MTBVAC, after each was given to 17 goats (2 months old) and then exposed for 9 months to goats infected with M. caprae. In general, MTBVAC-vaccinated goats showed higher interferon-gamma release than BCG vaccinated goats in response to bovine protein purified derivative and ESAT-6/CFP-10 antigens and the response was significantly higher than that observed in the control group until challenge. All animals showed lesions consistent with TB at the end of the study. Goats that received either vaccine showed significantly lower scores for pulmonary lymph nodes and total lesions than unvaccinated controls. Both MTBVAC and BCG vaccines proved to be immunogenic and effective in reducing severity of TB pathology caused by M. caprae. Our model system of natural TB transmission may be useful for evaluating and optimizing vaccines.


Asunto(s)
Vacuna BCG/inmunología , Enfermedades de las Cabras/inmunología , Inmunogenicidad Vacunal/inmunología , Mycoplasma/fisiología , Vacunas contra la Tuberculosis/inmunología , Tuberculosis/veterinaria , Animales , Enfermedades de las Cabras/transmisión , Cabras , Inmunidad Celular/efectos de los fármacos , Inmunidad Celular/inmunología , Inmunidad Humoral/efectos de los fármacos , Inmunidad Humoral/inmunología , Tuberculosis/inmunología , Tuberculosis/transmisión , Vacunas Atenuadas/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...