Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 7(1): 8853, 2017 08 18.
Article in English | MEDLINE | ID: mdl-28821804

ABSTRACT

During active TB in humans a spectrum of pulmonary granulomas with central necrosis and hypoxia exists. BALB/c mice, predominantly used in TB drug development, do not reproduce this complex pathology thereby inaccurately predicting clinical outcome. We found that Nos2 -/- mice incapable of NO-production in immune cells as microbial defence uniformly develop hypoxic necrotizing lung lesions, widely observed in human TB. To study the impact of hypoxic necrosis on the efficacy of antimycobacterials and drug candidates, we subjected Nos2 -/- mice with TB to monotherapy before or after establishment of human-like pathology. Isoniazid induced a drug-tolerant persister population only when necrotic lesions were present. Rifapentine was more potent than rifampin prior to development of human-like pathology and equally potent thereafter, in agreement with recent clinical trials. Pretomanid, delamanid and the pre-clinical candidate BTZ043 were bactericidal independent of pulmonary pathology. Linezolid was bacteriostatic in TB-infected Nos2 -/- mice but significantly improved lung pathology. Hypoxic necrotizing lesions rendered moxifloxacin less active. In conclusion, Nos2 -/- mice are a predictive TB drug development tool owing to their consistent development of human-like pathology.


Subject(s)
Hypoxia/metabolism , Necrosis/genetics , Necrosis/metabolism , Nitric Oxide Synthase Type II/deficiency , Tuberculosis, Pulmonary/etiology , Tuberculosis, Pulmonary/metabolism , Animals , Antitubercular Agents/pharmacology , Disease Models, Animal , Fibrosis , Foam Cells/immunology , Foam Cells/metabolism , Foam Cells/pathology , Humans , Hypoxia/pathology , Isoniazid/pharmacology , Mice , Mice, Knockout , Necrosis/pathology , Rifampin/analogs & derivatives , Rifampin/pharmacology , Treatment Outcome , Tuberculosis, Pulmonary/drug therapy , Tuberculosis, Pulmonary/pathology
2.
mBio ; 7(3)2016 05 24.
Article in English | MEDLINE | ID: mdl-27222470

ABSTRACT

UNLABELLED: The current tuberculosis (TB) vaccine, Mycobacterium bovis Bacillus Calmette-Guérin (BCG), provides insufficient protection against pulmonary TB. Previously, we generated a listeriolysin-expressing recombinant BCG strain, which to date has successfully completed phase I and phase IIa clinical trials. In an attempt to further improve efficacy, we deleted the antiapoptotic virulence gene nuoG, encoding NADH dehydrogenase 1 subunit G, from BCG ΔureC::hly In vitro, deletion of nuoG unexpectedly led to strongly increased recruitment of the autophagosome marker LC3 to the engulfed vaccine, suggesting that nuoG also affects xenophagic pathways. In mice, BCG ΔureC::hly ΔnuoG vaccination was safer than BCG and improved protection over that of parental BCG ΔureC::hly, significantly reducing TB load in murine lungs, ameliorating pulmonary pathology, and enhancing immune responses. Transcriptome analysis of draining lymph nodes after vaccination with either BCG ΔureC::hly or BCG ΔureC::hly ΔnuoG demonstrated earlier and stronger induction of immune responses than that with BCG SSI and suggested upregulation of inflammasome activation and interferon-induced GTPases. In summary, BCG ΔureC::hly ΔnuoG is a promising next-generation TB vaccine candidate with excellent efficacy and safety. IMPORTANCE: Autophagy and apoptosis are fundamental processes allowing cells to degrade their components or kill themselves, respectively. The immune system has adopted these mechanisms to eliminate intracellular pathogens. Residing in host cells, the causative agent of tuberculosis, Mycobacterium tuberculosis, has evolved strategies to set cellular programs of autophagy and apoptosis "on hold." The mycobacterial gene nuoG was found to prevent host cell apoptosis. We have deleted nuoG in the live vaccine candidate BCG ΔureC::hly, which is in phase II clinical development, to leave cellular apoptosis "on go" upon immunization. In preclinical models, this strategy boosted immunity and improved protection from M. tuberculosis infection. Unexpectedly, we obtained compelling evidence that mycobacterial nuoG facilitates inhibition of autophagic pathways, suggesting a new role for this gene in the host-pathogen interplay in tuberculosis.


Subject(s)
BCG Vaccine/genetics , BCG Vaccine/immunology , Electron Transport Complex I/genetics , Gene Deletion , Mycobacterium bovis/genetics , Mycobacterium bovis/immunology , Tuberculosis, Pulmonary/prevention & control , Animals , BCG Vaccine/administration & dosage , BCG Vaccine/adverse effects , Bacterial Load , Bacterial Proteins/genetics , Gene Expression Profiling , Inflammasomes/immunology , Lung/immunology , Lung/microbiology , Lung/pathology , Lymph Nodes/immunology , Lymph Nodes/microbiology , Mice , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/microbiology , Vaccination
3.
Microbes Infect ; 18(5): 364-8, 2016 05.
Article in English | MEDLINE | ID: mdl-26994939

ABSTRACT

The tuberculosis vaccine BCG ΔureC::hly is the most advanced BCG replacement candidate in phase II clinical development. Here we assess the protective capacity of the construct administered to mice as homologous prime-boost vaccine prior Mycobacterium tuberculosis infection and as post-exposure vaccine. Multiple immunization did not improve the superior protection of BCG ΔureC::hly over BCG. Animals with subclinical tuberculosis were better protected when vaccinated with BCG ΔureC::hly as compared to BCG. Our findings suggest further consideration of BCG ΔureC::hly as post-exposure vaccine.


Subject(s)
Asymptomatic Infections , BCG Vaccine/immunology , Bacterial Proteins/genetics , Gene Deletion , Mycobacterium bovis/immunology , Post-Exposure Prophylaxis/methods , Tuberculosis/prevention & control , Animals , BCG Vaccine/administration & dosage , Female , Mice, Inbred BALB C , Mycobacterium bovis/genetics , Treatment Outcome , Tuberculosis/immunology
4.
J Infect Dis ; 211(11): 1831-41, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25505299

ABSTRACT

BACKGROUND: The recombinant BCG ΔureC::hly (rBCG) vaccine candidate induces improved protection against tuberculosis over parental BCG (pBCG) in preclinical studies and has successfully completed a phase 2a clinical trial. However, the mechanisms responsible for the superior vaccine efficacy of rBCG are still incompletely understood. Here, we investigated the underlying biological mechanisms elicited by the rBCG vaccine candidate relevant to its protective efficacy. METHODS: THP-1 macrophages were infected with pBCG or rBCG, and inflammasome activation and autophagy were evaluated. In addition, mice were vaccinated with pBCG or rBCG, and gene expression in the draining lymph nodes was analyzed by microarray at day 1 after vaccination. RESULTS: BCG-derived DNA was detected in the cytosol of rBCG-infected macrophages. rBCG infection was associated with enhanced absent in melanoma 2 (AIM2) inflammasome activation, increased activation of caspases and production of interleukin (IL)-1ß and IL-18, as well as induction of AIM2-dependent and stimulator of interferon genes (STING)-dependent autophagy. Similarly, mice vaccinated with rBCG showed early increased expression of Il-1ß, Il-18, and Tmem173 (transmembrane protein 173; also known as STING). CONCLUSIONS: rBCG stimulates AIM2 inflammasome activation and autophagy, suggesting that these cell-autonomous functions should be exploited for improved vaccine design.


Subject(s)
Autophagy/immunology , BCG Vaccine/immunology , Inflammasomes/immunology , Tuberculosis/immunology , Vaccines, Synthetic/immunology , Animals , Cell Line , Female , Humans , Inflammation , Interleukin-18/immunology , Interleukin-1beta/immunology , Lymph Nodes/chemistry , Lymph Nodes/immunology , Mice , Mice, Inbred C57BL
5.
mBio ; 5(3): e01262-14, 2014 Jun 03.
Article in English | MEDLINE | ID: mdl-24895310

ABSTRACT

UNLABELLED: The only tuberculosis (TB) vaccine in use today, bacillus Calmette-Guérin (BCG), provides insufficient protection and can cause adverse events in immunocompromised individuals, such as BCGosis in HIV(+) newborns. We previously reported improved preclinical efficacy and safety of the recombinant vaccine candidate BCG ΔureC::hly, which secretes the pore-forming listeriolysin O of Listeria monocytogenes. Here, we evaluate a second-generation construct, BCG ΔureC::hly Δpdx1, which is deficient in pyridoxine synthase, an enzyme that is required for biosynthesis of the essential cofactor vitamin B6. This candidate was auxotrophic for vitamin B6 in a concentration-dependent manner, as was its survival in vivo. BCG ΔureC::hly Δpdx1 showed markedly restricted dissemination in subcutaneously vaccinated mice, which was ameliorated by dietary supplementation with vitamin B6. The construct was safer in severe combined immunodeficiency mice than the parental BCG ΔureC::hly. A prompt innate immune response to vaccination, measured by secretion of interleukin-6, granulocyte colony-stimulating factor, keratinocyte cytokine, and macrophage inflammatory protein-1α, remained independent of vitamin B6 administration, while acquired immunity, notably stimulation of antigen-specific CD4 T cells, B cells, and memory T cells, was contingent on vitamin B6 administration. The early protection provided by BCG ΔureC::hly Δpdx1 in a murine Mycobacterium tuberculosis aerosol challenge model consistently depended on vitamin B6 supplementation. Prime-boost vaccination increased protection against the canonical M. tuberculosis H37Rv laboratory strain and a clinical isolate of the Beijing/W lineage. We demonstrate that the efficacy of a profoundly attenuated recombinant BCG vaccine construct can be modulated by external administration of a small molecule. This principle fosters the development of safer vaccines required for immunocompromised individuals, notably HIV(+) infants. IMPORTANCE: Mycobacterium tuberculosis can synthesize the essential cofactor vitamin B6, while humans depend on dietary supplementation. Unlike the lipophilic vitamins A, D, and E, water-soluble vitamin B6 is well tolerated at high doses. We generated a vitamin B6 auxotroph of the phase II clinical tuberculosis vaccine candidate bacillus Calmette-Guérin ΔureC::hly. The next-generation candidate was profoundly attenuated compared to the parental strain. Adaptive immunity and protection in mice consistently depended on increased dietary vitamin B6 above the daily required dose. Control of vaccine efficacy via food supplements such as vitamin B6 could provide a fast track toward improved safety. Safer vaccines are urgently needed for HIV-infected individuals at high risk of adverse events in response to live vaccines.


Subject(s)
BCG Vaccine/immunology , Dietary Supplements , Mycobacterium bovis/immunology , Mycobacterium tuberculosis/immunology , Pyridoxine/administration & dosage , Tuberculosis/prevention & control , Vitamin B 6/biosynthesis , Animals , BCG Vaccine/genetics , Disease Models, Animal , Female , Genes, Bacterial , Immunity, Innate , Immunization, Secondary , Immunocompromised Host , Mice , Mutation , Mycobacterium bovis/genetics , Mycobacterium bovis/metabolism , Tuberculosis/mortality , Vaccination
6.
PLoS One ; 8(11): e78966, 2013.
Article in English | MEDLINE | ID: mdl-24236077

ABSTRACT

Bacillus Calmette-Guérin (BCG), the only approved tuberculosis vaccine, provides only limited protection. Previously, we generated a recombinant derivative (BCG ΔureC::hly), which secretes the pore-forming toxin listeriolysin O (LLO) of Listeria monocytogenes. This vaccine shows superior protection against tuberculosis in preclinical models and is safe in humans. Here we describe two new vaccine strains which express human interleukin-7 (hIL)-7 or hIL-18 in the genetic background of BCG ΔureC::hly to modulate specific T cell immunity. Both strains exhibited an uncompromised in vitro growth pattern, while inducing a proinflammatory cytokine profile in human dendritic cells (DCs). Human DCs harbouring either strain efficiently promoted secretion of IL-2 by autologous T cells in a coculture system, suggesting superior immunogenicity. BALB/c mice vaccinated with BCG ΔureC::hly, BCG ΔureC::hly_hIL7 or BCG ΔureC::hly_hIL18 developed a more robust Th1 response than after vaccination with parental BCG. Both strains provided significantly better protection than BCG in a murine Mycobacterium tuberculosis challenge model but efficacy remained comparable to that afforded by BCG ΔureC::hly. We conclude that expression of hIL-7 or hIL-18 enhanced specific T cell responses but failed to improve protection over BCG ΔureC::hly in mice.


Subject(s)
BCG Vaccine/immunology , Interleukin-18/biosynthesis , Interleukin-7/biosynthesis , Mycobacterium tuberculosis/immunology , Th1 Cells/immunology , Tuberculosis/prevention & control , Vaccination , Animals , Antigens, Bacterial/immunology , BCG Vaccine/genetics , CD40 Ligand/metabolism , Cells, Cultured , Female , Gene Expression , Humans , Interleukin-18/genetics , Interleukin-7/genetics , Lung/immunology , Lung/pathology , Mice , Mice, Inbred BALB C , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Spleen/immunology , Spleen/pathology , Th1 Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...