Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
Add more filters

Country/Region as subject
Publication year range
1.
PLoS Pathog ; 16(12): e1009061, 2020 12.
Article in English | MEDLINE | ID: mdl-33347499

ABSTRACT

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.


Subject(s)
Bacterial Proteins/genetics , Host Microbial Interactions/genetics , Mycobacterium tuberculosis/genetics , Protein Serine-Threonine Kinases/genetics , Animals , Bacterial Proteins/metabolism , Female , Genomics , Male , Mice , Mice, Inbred C57BL , Mycobacterium tuberculosis/pathogenicity , Polymorphism, Genetic/genetics , Protein Serine-Threonine Kinases/metabolism , Virulence/genetics
2.
PLoS Pathog ; 16(4): e1008404, 2020 04.
Article in English | MEDLINE | ID: mdl-32240273

ABSTRACT

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.


Subject(s)
Disease Models, Animal , Immunity, Innate/immunology , Monocytes/immunology , Mycobacterium tuberculosis/immunology , Pneumonia/prevention & control , Tuberculosis Vaccines/administration & dosage , Tuberculosis/prevention & control , Animals , BCG Vaccine/administration & dosage , BCG Vaccine/immunology , Cells, Cultured , Cellular Reprogramming , Female , Humans , Immunity, Innate/drug effects , Mice , Mice, Inbred C57BL , Monocytes/drug effects , Monocytes/metabolism , Mycobacterium tuberculosis/drug effects , Pneumonia/immunology , Pneumonia/microbiology , Tuberculosis/immunology , Tuberculosis/microbiology , Vaccination
3.
PLoS Genet ; 14(4): e1007282, 2018 04.
Article in English | MEDLINE | ID: mdl-29649213

ABSTRACT

The insertion Sequence IS6110, only present in the pathogens of the Mycobacterium tuberculosis Complex (MTBC), has been the gold-standard epidemiological marker for TB for more than 25 years, but biological implications of IS6110 transposition during MTBC adaptation to humans remain elusive. By studying 2,236 clinical isolates typed by IS6110-RFLP and covering the MTBC, we remarked a lineage-specific content of IS6110 being higher in modern globally distributed strains. Once observed the IS6110 distribution in the MTBC, we selected representative isolates and found a correlation between the normalized expression of IS6110 and its abundance in MTBC chromosomes. We also studied the molecular regulation of IS6110 transposition and we found a synergistic action of two post-transcriptional mechanisms: a -1 ribosomal frameshift and a RNA pseudoknot which interferes translation. The construction of a transcriptionally active transposase resulted in 20-fold increase of the transposition frequency. Finally, we examined transposition in M. bovis and M. tuberculosis during laboratory starvation and in a mouse infection model of TB. Our results shown a higher transposition in M. tuberculosis, that preferably happens during TB infection in mice and after one year of laboratory culture, suggesting that IS6110 transposition is dynamically adapted to the host and to adverse growth conditions.


Subject(s)
DNA Transposable Elements/genetics , Mycobacterium tuberculosis/genetics , Animals , DNA Copy Number Variations , Disease Models, Animal , Frameshifting, Ribosomal , Genes, Bacterial , Humans , Mice , Mycobacterium tuberculosis/growth & development , RNA Processing, Post-Transcriptional , Tuberculosis/microbiology
4.
Vet Res ; 50(1): 82, 2019 Oct 15.
Article in English | MEDLINE | ID: mdl-31615555

ABSTRACT

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.


Subject(s)
BCG Vaccine/immunology , Goat Diseases/immunology , Immunogenicity, Vaccine/immunology , Mycoplasma/physiology , Tuberculosis Vaccines/immunology , Tuberculosis/veterinary , Animals , Goat Diseases/transmission , Goats , Immunity, Cellular/drug effects , Immunity, Cellular/immunology , Immunity, Humoral/drug effects , Immunity, Humoral/immunology , Tuberculosis/immunology , Tuberculosis/transmission , Vaccines, Attenuated/immunology
5.
J Infect Dis ; 213(5): 831-9, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26494773

ABSTRACT

Some of the most promising novel tuberculosis vaccine strategies currently under development are based on respiratory vaccination, mimicking the natural route of infection. In this work, we have compared pulmonary and subcutaneous delivery of BCG vaccine in the tuberculosis-susceptible DBA/2 mouse strain, a model in which parenterally administered BCG vaccine does not protect against tuberculosis. Our data show that intranasally but not subcutaneously administered BCG confers robust protection against pulmonary tuberculosis challenge. In addition, our results indicate that pulmonary vaccination triggers a Mycobacterium tuberculosis-specific mucosal immune response orchestrated by interleukin 17A (IL-17A). Thus, IL-17A neutralization in vivo reduces protection and abrogates M. tuberculosis-specific immunoglobulin A (IgA) secretion to respiratory airways and lung expression of polymeric immunoglobulin receptor induced following intranasal vaccination. Together, our results demonstrate that pulmonary delivery of BCG can overcome the lack of protection observed when BCG is given parenterally, suggesting that respiratory tuberculosis vaccines could have an advantage in tuberculosis-endemic countries, where intradermally administered BCG has inefficient effectiveness against pulmonary tuberculosis.


Subject(s)
BCG Vaccine/immunology , Interleukin-17/metabolism , Tuberculosis/prevention & control , Administration, Intranasal , Animals , BCG Vaccine/administration & dosage , Female , Immunoglobulin A/metabolism , Injections, Subcutaneous , Interleukin-17/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/metabolism
6.
J Biol Chem ; 290(11): 6868-77, 2015 Mar 13.
Article in English | MEDLINE | ID: mdl-25605735

ABSTRACT

Cytotoxic T cells (Tc) use perforin and granzyme B (gzmB) to kill virus-infected cells and cancer cells. Recent evidence suggests that human gzmB primarily induces apoptosis via the intrinsic mitochondrial pathway by either cleaving Bid or activating Bim leading to the activation of Bak/Bax and subsequent generation of active caspase-3. In contrast, mouse gzmB is thought to predominantly induce apoptosis by directly processing pro-caspase-3. However, in certain mouse cell types gzmB-mediated apoptosis mainly occurs via the mitochondrial pathway. To investigate whether Bim is involved under the latter conditions, we have now employed ex vivo virus-immune mouse Tc that selectively kill by using perforin and gzmB (gzmB(+)Tc) as effector cells and wild type as well as Bim- or Bak/Bax-deficient spontaneously (3T9) or virus-(SV40) transformed mouse embryonic fibroblast cells as targets. We show that gzmB(+)Tc-mediated apoptosis (phosphatidylserine translocation, mitochondrial depolarization, cytochrome c release, and caspase-3 activation) was severely reduced in 3T9 cells lacking either Bim or both Bak and Bax. This outcome was related to the ability of Tc cells to induce the degradation of Mcl-1 and Bcl-XL, the anti-apoptotic counterparts of Bim. In contrast, gzmB(+)Tc-mediated apoptosis was not affected in SV40-transformed mouse embryonic fibroblast cells lacking Bak/Bax. The data provide evidence that Bim participates in mouse gzmB(+)Tc-mediated apoptosis of certain targets by activating the mitochondrial pathway and suggest that the mode of cell death depends on the target cell. Our results suggest that the various molecular events leading to transformation and/or immortalization of cells have an impact on their relative resistance to the multiple gzmB(+)Tc-induced death pathways.


Subject(s)
Apoptosis Regulatory Proteins/immunology , Apoptosis , Granzymes/immunology , Membrane Proteins/immunology , Mitochondria/immunology , Proto-Oncogene Proteins/immunology , T-Lymphocytes, Cytotoxic/enzymology , Animals , Apoptosis Regulatory Proteins/genetics , Bcl-2-Like Protein 11 , Caspase 3/immunology , Cell Line , Fibroblasts/cytology , Fibroblasts/immunology , Fibroblasts/metabolism , Gene Deletion , Immunotherapy , Membrane Proteins/genetics , Mice , Myeloid Cell Leukemia Sequence 1 Protein/immunology , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Neoplasms/therapy , Pore Forming Cytotoxic Proteins/immunology , Proteolysis , Proto-Oncogene Proteins/genetics , T-Lymphocytes, Cytotoxic/immunology
7.
Cell Microbiol ; 17(11): 1537-60, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26045209

ABSTRACT

Klebsiella pneumoniae is an important cause of community-acquired and nosocomial pneumonia. Evidence indicates that Klebsiella might be able to persist intracellularly within a vacuolar compartment. This study was designed to investigate the interaction between Klebsiella and macrophages. Engulfment of K. pneumoniae was dependent on host cytoskeleton, cell plasma membrane lipid rafts and the activation of phosphoinositide 3-kinase (PI3K). Microscopy studies revealed that K. pneumoniae resides within a vacuolar compartment, the Klebsiella-containing vacuole (KCV), which traffics within vacuoles associated with the endocytic pathway. In contrast to UV-killed bacteria, the majority of live bacteria did not co-localize with markers of the lysosomal compartment. Our data suggest that K. pneumoniae triggers a programmed cell death in macrophages displaying features of apoptosis. Our efforts to identify the mechanism(s) whereby K. pneumoniae prevents the fusion of the lysosomes to the KCV uncovered the central role of the PI3K-Akt-Rab14 axis to control the phagosome maturation. Our data revealed that the capsule is dispensable for Klebsiella intracellular survival if bacteria were not opsonized. Furthermore, the environment found by Klebsiella within the KCV triggered the down-regulation of the expression of cps. Altogether, this study proves evidence that K. pneumoniae survives killing by macrophages by manipulating phagosome maturation that may contribute to Klebsiella pathogenesis.


Subject(s)
Klebsiella pneumoniae/immunology , Klebsiella pneumoniae/physiology , Lysosomes/metabolism , Macrophages/microbiology , Microbial Viability , Animals , Cells, Cultured , Host-Pathogen Interactions , Humans , Macrophages/immunology , Macrophages/pathology , Mice , Vacuoles/microbiology
8.
Infect Immun ; 82(8): 3446-56, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24891105

ABSTRACT

The ESX-1 secreted virulence factor ESAT-6 is one of the major and most well-studied virulence factors of Mycobacterium tuberculosis, given that its inactivation severely attenuates virulent mycobacteria. In this work, we show that clinical isolates of M. tuberculosis produce and secrete larger amounts of ESAT-6 than the widely used M. tuberculosis H37Rv laboratory strain. A search for the genetic polymorphisms underlying this observation showed that whiB6 (rv3862c), a gene upstream of the ESX-1 genetic locus that has not previously been found to be implicated in the regulation of the ESX-1 secretory apparatus, presents a unique single nucleotide insertion in its promoter region in strains H37Rv and H37Ra. This polymorphism is not present in any of the other publicly available M. tuberculosis complex genomes or in any of the 76 clinical M. tuberculosis isolates analyzed in our laboratory. We demonstrate that in consequence, the virulence master regulator PhoP downregulates whiB6 expression in H37Rv, while it upregulates its expression in clinical strains. Importantly, reintroduction of the wild-type (WT) copy of whiB6 in H37Rv restored ESAT-6 production and secretion to the level of clinical strains. Hence, we provide clear evidence that in M. tuberculosis--with the exception of the H37Rv strain--ESX-1 expression is regulated by WhiB6 as part of the PhoP regulon, which adds another level of complexity to the regulation of ESAT-6 secretion with a potential role in virulence adaptation.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Secretion Systems , Gene Expression Regulation, Bacterial , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism , Polymorphism, Single Nucleotide , Antigens, Bacterial , Promoter Regions, Genetic , Regulon
10.
Sci Rep ; 14(1): 13133, 2024 06 07.
Article in English | MEDLINE | ID: mdl-38849432

ABSTRACT

The short-lived nature and heterogeneity of Natural Killer (NK) cells limit the development of NK cell-based therapies, despite their proven safety and efficacy against cancer. Here, we describe the biological basis, detailed phenotype and function of long-lived anti-tumour human NK cells (CD56highCD16+), obtained without cell sorting or feeder cells, after priming of peripheral blood cells with Bacillus Calmette-Guérin (BCG). Further, we demonstrate that survival doses of a cytokine combination, excluding IL18, administered just weekly to BCG-primed NK cells avoids innate lymphocyte exhaustion and leads to specific long-term proliferation of innate cells that exert potent cytotoxic function against a broad range of solid tumours, mainly through NKG2D. Strikingly, a NKG2C+CD57-FcεRIγ+ NK cell population expands after BCG and cytokine stimulation, independently of HCMV serology. This strategy was exploited to rescue anti-tumour NK cells even from the suppressor environment of cancer patients' bone marrow, demonstrating that BCG confers durable anti-tumour features to NK cells.


Subject(s)
Cell Proliferation , Killer Cells, Natural , Killer Cells, Natural/immunology , Killer Cells, Natural/drug effects , Humans , Cell Proliferation/drug effects , Neoplasms/immunology , Neoplasms/drug therapy , BCG Vaccine/immunology , BCG Vaccine/administration & dosage , Mycobacterium bovis/immunology , Lymphocyte Activation/drug effects , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Interleukins/metabolism , CD56 Antigen/metabolism , NK Cell Lectin-Like Receptor Subfamily C/metabolism
11.
iScience ; 27(2): 108869, 2024 Feb 16.
Article in English | MEDLINE | ID: mdl-38318361

ABSTRACT

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.

12.
Front Immunol ; 15: 1387454, 2024.
Article in English | MEDLINE | ID: mdl-38799468

ABSTRACT

Introduction: Mycobacteria are known to exert a range of heterologous effects on the immune system. The mycobacteria-based Freund's Complete Adjuvant is a potent non-specific stimulator of the immune response used in immunization protocols promoting antibody production, and Mycobacterium bovis Bacille Calmette Guérin (BCG) vaccination has been linked with decreased morbidity and mortality beyond the specific protection it provides against tuberculosis (TB) in some populations and age groups. The role of heterologous antibodies in this phenomenon, if any, remains unclear and under-studied. Methods: We set out to evaluate antibody responses to a range of unrelated pathogens following infection with Mycobacterium tuberculosis (M.tb) and vaccination with BCG or a candidate TB vaccine, MTBVAC, in non-human primates. Results: We demonstrate a significant increase in the titer of antibodies against SARS-CoV-2, cytomegalovirus, Epstein-Barr virus, tetanus toxoid, and respiratory syncytial virus antigens following low-dose aerosol infection with M.tb. The magnitude of some of these responses correlated with TB disease severity. However, vaccination with BCG administered by the intradermal, intravenous or aerosol routes, or intradermal delivery of MTBVAC, did not increase antibody responses against unrelated pathogens. Discussion: Our findings suggest that it is unlikely that heterologous antibodies contribute to the non-specific effects of these vaccines. The apparent dysregulation of B cell responses associated with TB disease warrants further investigation, with potential implications for risk of B cell cancers and novel therapeutic strategies.


Subject(s)
BCG Vaccine , Mycobacterium tuberculosis , Tuberculosis , Vaccination , Animals , BCG Vaccine/immunology , BCG Vaccine/administration & dosage , Tuberculosis/immunology , Tuberculosis/prevention & control , Mycobacterium tuberculosis/immunology , Antibodies, Bacterial/immunology , Antibodies, Bacterial/blood , Antibodies, Viral/immunology , Antibodies, Viral/blood , Tuberculosis Vaccines/immunology , Tuberculosis Vaccines/administration & dosage , Female , Macaca mulatta , SARS-CoV-2/immunology , COVID-19/immunology , COVID-19/prevention & control , Immunity, Heterologous , Male
13.
Oncoimmunology ; 12(1): 2160094, 2023.
Article in English | MEDLINE | ID: mdl-36567803

ABSTRACT

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.


Subject(s)
Antineoplastic Agents , Mycobacterium bovis , Urinary Bladder Neoplasms , Humans , BCG Vaccine/therapeutic use , Leukocytes, Mononuclear , Killer Cells, Natural , Urinary Bladder Neoplasms/therapy , T-Lymphocytes
14.
Nat Commun ; 14(1): 6090, 2023 10 04.
Article in English | MEDLINE | ID: mdl-37794033

ABSTRACT

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.


Subject(s)
Lung Neoplasms , Urinary Bladder Neoplasms , Mice , Animals , BCG Vaccine , Urinary Bladder Neoplasms/pathology , CD8-Positive T-Lymphocytes , Administration, Intravenous , Immunity, Cellular , Killer Cells, Natural , Lung/pathology , Lung Neoplasms/drug therapy
15.
Mol Ther Nucleic Acids ; 27: 1235-1248, 2022 Mar 08.
Article in English | MEDLINE | ID: mdl-35282413

ABSTRACT

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.

16.
J Immunother Cancer ; 10(7)2022 07.
Article in English | MEDLINE | ID: mdl-35781395

ABSTRACT

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.


Subject(s)
Urinary Bladder Neoplasms , Animals , BCG Vaccine/therapeutic use , Bacteria , Immune Checkpoint Inhibitors , Immunotherapy , Mice , Urinary Bladder Neoplasms/therapy
17.
Vaccine ; 39(50): 7277-7285, 2021 12 08.
Article in English | MEDLINE | ID: mdl-34238608

ABSTRACT

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.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis Vaccines , Tuberculosis , BCG Vaccine , Humans , Infant, Newborn , Tuberculosis/prevention & control , Vaccines, Attenuated
18.
EBioMedicine ; 64: 103186, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33478923

ABSTRACT

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].


Subject(s)
Asthma/immunology , Asthma/therapy , Tuberculosis Vaccines/therapeutic use , Vaccines, Attenuated/therapeutic use , Administration, Intranasal , Airway Remodeling/immunology , Allergens/immunology , Animals , BCG Vaccine , Biomarkers , Cellular Microenvironment/immunology , Cytokines/metabolism , Disease Models, Animal , Eosinophils/immunology , Eosinophils/metabolism , Eosinophils/pathology , Female , Immunization , Mice , Ovalbumin/immunology , Tuberculosis Vaccines/administration & dosage , Vaccines, Attenuated/administration & dosage
19.
Front Immunol ; 12: 622995, 2021.
Article in English | MEDLINE | ID: mdl-33708215

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/immunology , BCG Vaccine/immunology , Cell Degranulation , Immunotherapy , Killer Cells, Natural/immunology , Lymphocyte Activation , Mycobacterium bovis/immunology , Urinary Bladder Neoplasms/therapy , BCG Vaccine/genetics , CD3 Complex/metabolism , CD56 Antigen/metabolism , Cell Proliferation , Coculture Techniques , Humans , K562 Cells , Killer Cells, Natural/metabolism , Mycobacterium bovis/genetics , Tumor Microenvironment , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/metabolism , Vaccines, Inactivated/genetics , Vaccines, Inactivated/immunology
20.
EBioMedicine ; 65: 103254, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33711798

ABSTRACT

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".


Subject(s)
BCG Vaccine/administration & dosage , Diphtheria-Tetanus-acellular Pertussis Vaccines/administration & dosage , Immunity, Humoral , Whooping Cough/prevention & control , Animals , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , Diphtheria/immunology , Diphtheria/prevention & control , Disease Models, Animal , Female , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Incidence , Interferon-gamma/metabolism , Mice , Mice, Inbred BALB C , Tetanus/immunology , Tetanus/prevention & control , Th1 Cells/cytology , Th1 Cells/immunology , Th1 Cells/metabolism , Vaccination , Whooping Cough/epidemiology , Whooping Cough/immunology
SELECTION OF CITATIONS
SEARCH DETAIL