Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Antioxidants (Basel) ; 10(8)2021 Aug 13.
Article in English | MEDLINE | ID: mdl-34439535

ABSTRACT

H2S is a potent gasotransmitter in eukaryotes and bacteria. Host-derived H2S has been shown to profoundly alter M. tuberculosis (Mtb) energy metabolism and growth. However, compelling evidence for endogenous production of H2S and its role in Mtb physiology is lacking. We show that multidrug-resistant and drug-susceptible clinical Mtb strains produce H2S, whereas H2S production in non-pathogenic M. smegmatis is barely detectable. We identified Rv3684 (Cds1) as an H2S-producing enzyme in Mtb and show that cds1 disruption reduces, but does not eliminate, H2S production, suggesting the involvement of multiple genes in H2S production. We identified endogenous H2S to be an effector molecule that maintains bioenergetic homeostasis by stimulating respiration primarily via cytochrome bd. Importantly, H2S plays a key role in central metabolism by modulating the balance between oxidative phosphorylation and glycolysis, and it functions as a sink to recycle sulfur atoms back to cysteine to maintain sulfur homeostasis. Lastly, Mtb-generated H2S regulates redox homeostasis and susceptibility to anti-TB drugs clofazimine and rifampicin. These findings reveal previously unknown facets of Mtb physiology and have implications for routine laboratory culturing, understanding drug susceptibility, and improved diagnostics.

2.
Front Cell Infect Microbiol ; 10: 586923, 2020.
Article in English | MEDLINE | ID: mdl-33330130

ABSTRACT

For centuries, hydrogen sulfide (H2S) was considered primarily as a poisonous gas and environmental hazard. However, with the discovery of prokaryotic and eukaryotic enzymes for H2S production, breakdown, and utilization, H2S has emerged as an important signaling molecule in a wide range of physiological and pathological processes. Hence, H2S is considered a gasotransmitter along with nitric oxide (•NO) and carbon monoxide (CO). Surprisingly, despite having overlapping functions with •NO and CO, the role of host H2S in microbial pathogenesis is understudied and represents a gap in our knowledge. Given the numerous reports that followed the discovery of •NO and CO and their respective roles in microbial pathogenesis, we anticipate a rapid increase in studies that further define the importance of H2S in microbial pathogenesis, which may lead to new virulence paradigms. Therefore, this review provides an overview of sulfide chemistry, enzymatic production of H2S, and the importance of H2S in metabolism and immunity in response to microbial pathogens. We then describe our current understanding of the role of host-derived H2S in tuberculosis (TB) disease, including its influences on host immunity and bioenergetics, and on Mycobacterium tuberculosis (Mtb) growth and survival. Finally, this review discusses the utility of H2S-donor compounds, inhibitors of H2S-producing enzymes, and their potential clinical significance.


Subject(s)
Hydrogen Sulfide , Mycobacterium tuberculosis , Tuberculosis , Carbon Monoxide , Humans , Nitric Oxide
3.
Proc Natl Acad Sci U S A ; 117(12): 6663-6674, 2020 03 24.
Article in English | MEDLINE | ID: mdl-32139610

ABSTRACT

The ubiquitous gasotransmitter hydrogen sulfide (H2S) has been recognized to play a crucial role in human health. Using cystathionine γ-lyase (CSE)-deficient mice, we demonstrate an unexpected role of H2S in Mycobacterium tuberculosis (Mtb) pathogenesis. We showed that Mtb-infected CSE-/- mice survive longer than WT mice, and support reduced pathology and lower bacterial burdens in the lung, spleen, and liver. Similarly, in vitro Mtb infection of macrophages resulted in reduced colony forming units in CSE-/- cells. Chemical complementation of infected WT and CSE-/- macrophages using the slow H2S releaser GYY3147 and the CSE inhibitor DL-propargylglycine demonstrated that H2S is the effector molecule regulating Mtb survival in macrophages. Furthermore, we demonstrate that CSE promotes an excessive innate immune response, suppresses the adaptive immune response, and reduces circulating IL-1ß, IL-6, TNF-α, and IFN-γ levels in response to Mtb infection. Notably, Mtb infected CSE-/- macrophages show increased flux through glycolysis and the pentose phosphate pathway, thereby establishing a critical link between H2S and central metabolism. Our data suggest that excessive H2S produced by the infected WT mice reduce HIF-1α levels, thereby suppressing glycolysis and production of IL-1ß, IL-6, and IL-12, and increasing bacterial burden. Clinical relevance was demonstrated by the spatial distribution of H2S-producing enzymes in human necrotic, nonnecrotic, and cavitary pulmonary tuberculosis (TB) lesions. In summary, CSE exacerbates TB pathogenesis by altering immunometabolism in mice and inhibiting CSE or modulating glycolysis are potential targets for host-directed TB control.


Subject(s)
Carbon/metabolism , Cystathionine gamma-Lyase/physiology , Hydrogen Sulfide/toxicity , Mycobacterium tuberculosis/immunology , Tuberculosis, Pulmonary/etiology , Alkynes/pharmacology , Animals , Cystathionine gamma-Lyase/antagonists & inhibitors , Cytokines/metabolism , Enzyme Inhibitors/pharmacology , Glycine/analogs & derivatives , Glycine/pharmacology , Glycolysis , Hydrogen Sulfide/metabolism , Lymphocytes/drug effects , Lymphocytes/immunology , Lymphocytes/metabolism , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mycobacterium tuberculosis/drug effects , Myeloid Cells/drug effects , Myeloid Cells/immunology , Myeloid Cells/metabolism , Signal Transduction , Tuberculosis, Pulmonary/metabolism , Tuberculosis, Pulmonary/pathology
4.
Nat Commun ; 11(1): 557, 2020 Jan 28.
Article in English | MEDLINE | ID: mdl-31992699

ABSTRACT

Hydrogen sulfide (H2S) is involved in numerous pathophysiological processes and shares overlapping functions with CO and •NO. However, the importance of host-derived H2S in microbial pathogenesis is unknown. Here we show that Mtb-infected mice deficient in the H2S-producing enzyme cystathionine ß-synthase (CBS) survive longer with reduced organ burden, and that pharmacological inhibition of CBS reduces Mtb bacillary load in mice. High-resolution respirometry, transcriptomics and mass spectrometry establish that H2S stimulates Mtb respiration and bioenergetics predominantly via cytochrome bd oxidase, and that H2S reverses •NO-mediated inhibition of Mtb respiration. Further, exposure of Mtb to H2S regulates genes involved in sulfur and copper metabolism and the Dos regulon. Our results indicate that Mtb exploits host-derived H2S to promote growth and disease, and suggest that host-directed therapies targeting H2S production may be potentially useful for the management of tuberculosis and other microbial infections.


Subject(s)
Hydrogen Sulfide/pharmacology , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/metabolism , Mycobacterium tuberculosis/pathogenicity , Animals , Copper/metabolism , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/metabolism , Cytokines/blood , Disease Models, Animal , Electron Transport Complex IV/metabolism , Energy Metabolism , Female , Gene Expression Regulation, Bacterial/drug effects , Homeostasis , Lung/pathology , Macrophages , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mycobacterium tuberculosis/genetics , RAW 264.7 Cells , Regulon , Sulfur/metabolism , Transcriptome , Tuberculosis
6.
Cell Rep ; 25(7): 1938-1952.e5, 2018 11 13.
Article in English | MEDLINE | ID: mdl-30428359

ABSTRACT

Heme oxygenase-1 (HO-1) is a cytoprotective enzyme that controls inflammatory responses and redox homeostasis; however, its role during pulmonary tuberculosis (TB) remains unclear. Using freshly resected human TB lung tissue, we examined the role of HO-1 within the cellular and pathological spectrum of TB. Flow cytometry and histopathological analysis of human TB lung tissues showed that HO-1 is expressed primarily in myeloid cells and that HO-1 levels in these cells were directly proportional to cytoprotection. HO-1 mitigates TB pathophysiology by diminishing myeloid cell-mediated oxidative damage caused by reactive oxygen and/or nitrogen intermediates, which control granulocytic karyorrhexis to generate a zonal HO-1 response. Using whole-body or myeloid-specific HO-1-deficient mice, we demonstrate that HO-1 is required to control myeloid cell infiltration and inflammation to protect against TB progression. Overall, this study reveals that zonation of HO-1 in myeloid cells modulates free-radical-mediated stress, which regulates human TB immunopathology.


Subject(s)
Free Radicals/metabolism , Heme Oxygenase-1/metabolism , Tuberculosis/immunology , Tuberculosis/pathology , Animals , Arginase/metabolism , CD4-Positive T-Lymphocytes/immunology , Cytokines/metabolism , Granuloma/pathology , Heme Oxygenase-1/deficiency , Humans , Inflammation/pathology , Lung/pathology , Mice, Inbred C57BL , Mice, Knockout , Mycobacterium tuberculosis/physiology , Myeloid Cells/enzymology , NF-E2-Related Factor 2/metabolism , Neutrophils/metabolism , Nitric Oxide Synthase Type II/metabolism , Tuberculosis/enzymology , Tuberculosis/microbiology
7.
Front Immunol ; 9: 860, 2018.
Article in English | MEDLINE | ID: mdl-29774023

ABSTRACT

Iron is an essential factor for the growth and virulence of Mycobacterium tuberculosis (Mtb). However, little is known about the mechanisms by which the host controls iron availability during infection. Since ferritin heavy chain (FtH) is a major intracellular source of reserve iron in the host, we hypothesized that the lack of FtH would cause dysregulated iron homeostasis to exacerbate TB disease. Therefore, we used knockout mice lacking FtH in myeloid-derived cell populations to study Mtb disease progression. We found that FtH plays a critical role in protecting mice against Mtb, as evidenced by increased organ burden, extrapulmonary dissemination, and decreased survival in Fth-/- mice. Flow cytometry analysis showed that reduced levels of FtH contribute to an excessive inflammatory response to exacerbate disease. Extracellular flux analysis showed that FtH is essential for maintaining bioenergetic homeostasis through oxidative phosphorylation. In support of these findings, RNAseq and mass spectrometry analyses demonstrated an essential role for FtH in mitochondrial function and maintenance of central intermediary metabolism in vivo. Further, we show that FtH deficiency leads to iron dysregulation through the hepcidin-ferroportin axis during infection. To assess the clinical significance of our animal studies, we performed a clinicopathological analysis of iron distribution within human TB lung tissue and showed that Mtb severely disrupts iron homeostasis in distinct microanatomic locations of the human lung. We identified hemorrhage as a major source of metabolically inert iron deposition. Importantly, we observed increased iron levels in human TB lung tissue compared to healthy tissue. Overall, these findings advance our understanding of the link between iron-dependent energy metabolism and immunity and provide new insight into iron distribution within the spectrum of human pulmonary TB. These metabolic mechanisms could serve as the foundation for novel host-directed strategies.


Subject(s)
Apoferritins/immunology , Iron/metabolism , Lung/pathology , Mycobacterium tuberculosis/immunology , Tuberculosis, Pulmonary/immunology , Animals , Apoferritins/genetics , Apoferritins/metabolism , Case-Control Studies , Disease Models, Animal , Disease Susceptibility/immunology , Disease Susceptibility/microbiology , Energy Metabolism/immunology , Female , Ferritins , Healthy Volunteers , Hepcidins/metabolism , Humans , Iron/analysis , Iron/immunology , Lung/microbiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidoreductases , Tuberculosis, Pulmonary/microbiology , Tuberculosis, Pulmonary/pathology
9.
PLoS Pathog ; 13(5): e1006389, 2017 May.
Article in English | MEDLINE | ID: mdl-28542477

ABSTRACT

Signals modulating the production of Mycobacterium tuberculosis (Mtb) virulence factors essential for establishing long-term persistent infection are unknown. The WhiB3 redox regulator is known to regulate the production of Mtb virulence factors, however the mechanisms of this modulation are unknown. To advance our understanding of the mechanisms involved in WhiB3 regulation, we performed Mtb in vitro, intraphagosomal and infected host expression analyses. Our Mtb expression analyses in conjunction with extracellular flux analyses demonstrated that WhiB3 maintains bioenergetic homeostasis in response to available carbon sources found in vivo to establish Mtb infection. Our infected host expression analysis indicated that WhiB3 is involved in regulation of the host cell cycle. Detailed cell-cycle analysis revealed that Mtb infection inhibited the macrophage G1/S transition, and polyketides under WhiB3 control arrested the macrophages in the G0-G1 phase. Notably, infection with the Mtb whiB3 mutant or polyketide mutants had little effect on the macrophage cell cycle and emulated the uninfected cells. This suggests that polyketides regulated by Mtb WhiB3 are responsible for the cell cycle arrest observed in macrophages infected with the wild type Mtb. Thus, our findings demonstrate that Mtb WhiB3 maintains bioenergetic homeostasis to produce polyketide and lipid cyclomodulins that target the host cell cycle. This is a new mechanism whereby Mtb modulates the immune system by altering the host cell cycle to promote long-term persistence. This new knowledge could serve as the foundation for new host-directed therapeutic discovery efforts that target the host cell cycle.


Subject(s)
Mycobacterium tuberculosis/physiology , Tuberculosis/physiopathology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Female , G1 Phase Cell Cycle Checkpoints , Host-Pathogen Interactions , Humans , Macrophages/metabolism , Macrophages/microbiology , Mice, Inbred BALB C , Mycobacterium tuberculosis/genetics , S Phase Cell Cycle Checkpoints , Transcription Factors/genetics , Transcription Factors/metabolism , Tuberculosis/metabolism , Tuberculosis/microbiology
10.
Nat Commun ; 7: 12393, 2016 08 10.
Article in English | MEDLINE | ID: mdl-27506290

ABSTRACT

The Mycobacterium tuberculosis (Mtb) electron transport chain (ETC) has received significant attention as a drug target, however its vulnerability may be affected by its flexibility in response to disruption. Here we determine the effect of the ETC inhibitors bedaquiline, Q203 and clofazimine on the Mtb ETC, and the value of the ETC as a drug target, by measuring Mtb's respiration using extracellular flux technology. We find that Mtb's ETC rapidly reroutes around inhibition by these drugs and increases total respiration to maintain ATP levels. Rerouting is possible because Mtb rapidly switches between terminal oxidases, and, unlike eukaryotes, is not susceptible to back pressure. Increased ETC activity potentiates clofazimine's production of reactive oxygen species, causing rapid killing in vitro and in a macrophage model. Our results indicate that combination therapy targeting the ETC can be exploited to enhance killing of Mtb.


Subject(s)
Antitubercular Agents/pharmacology , Electron Transport Chain Complex Proteins/antagonists & inhibitors , Mycobacterium tuberculosis/physiology , Reactive Oxygen Species/metabolism , Tuberculosis, Multidrug-Resistant/drug therapy , Adenosine Triphosphate/metabolism , Animals , Antitubercular Agents/therapeutic use , Clofazimine/pharmacology , Clofazimine/therapeutic use , Diarylquinolines/pharmacology , Diarylquinolines/therapeutic use , Drug Therapy, Combination/methods , Hep G2 Cells , Humans , Imidazoles/chemical synthesis , Imidazoles/pharmacology , Imidazoles/therapeutic use , Inhibitory Concentration 50 , Macrophages/microbiology , Mice , Mutation , Mycobacterium tuberculosis/drug effects , Piperidines/chemical synthesis , Piperidines/pharmacology , Piperidines/therapeutic use , Pyridines/chemical synthesis , Pyridines/pharmacology , Pyridines/therapeutic use , RAW 264.7 Cells , Tuberculosis, Multidrug-Resistant/microbiology
11.
Nitric Oxide ; 59: 28-41, 2016 09 30.
Article in English | MEDLINE | ID: mdl-27387335

ABSTRACT

Mycobacterium tuberculosis (Mtb) is a facultative intracellular pathogen and the second largest contributor to global mortality caused by an infectious agent after HIV. In infected host cells, Mtb is faced with a harsh intracellular environment including hypoxia and the release of nitric oxide (NO) and carbon monoxide (CO) by immune cells. Hypoxia, NO and CO induce a state of in vitro dormancy where Mtb senses these gases via the DosS and DosT heme sensor kinase proteins, which in turn induce a set of ∼47 genes, known as the Mtb Dos dormancy regulon. On the contrary, both iNOS and HO-1, which produce NO and CO, respectively, have been shown to be important against mycobacterial disease progression. In this review, we discuss the impact of O2, NO and CO on Mtb physiology and in host responses to Mtb infection as well as the potential role of another major endogenous gas, hydrogen sulfide (H2S), in Mtb pathogenesis.


Subject(s)
Gasotransmitters/physiology , Mycobacterium tuberculosis/physiology , Tuberculosis, Pulmonary/metabolism , Carbon Monoxide/physiology , Humans , Hydrogen Sulfide/metabolism , Mycobacterium tuberculosis/genetics , Nitric Oxide/physiology , Oxygen/physiology , Reactive Oxygen Species/metabolism , Tuberculosis, Pulmonary/microbiology
12.
J Biol Chem ; 290(23): 14407-17, 2015 Jun 05.
Article in English | MEDLINE | ID: mdl-25847237

ABSTRACT

Mycobacterium tuberculosis, the causative agent of tuberculosis, is an ancient pathogen and a major cause of death worldwide. Although various virulence factors of M. tuberculosis have been identified, its pathogenesis remains incompletely understood. TlyA is a virulence factor in several bacterial infections and is evolutionarily conserved in many Gram-positive bacteria, but its function in M. tuberculosis pathogenesis has not been elucidated. Here, we report that TlyA significantly contributes to the pathogenesis of M. tuberculosis. We show that a TlyA mutant M. tuberculosis strain induces increased IL-12 and reduced IL-1ß and IL-10 cytokine responses, which sharply contrasts with the immune responses induced by wild type M. tuberculosis. Furthermore, compared with wild type M. tuberculosis, TlyA-deficient M. tuberculosis bacteria are more susceptible to autophagy in macrophages. Consequently, animals infected with the TlyA mutant M. tuberculosis organisms exhibited increased host-protective immune responses, reduced bacillary load, and increased survival compared with animals infected with wild type M. tuberculosis. Thus, M. tuberculosis employs TlyA as a host evasion factor, thereby contributing to its virulence.


Subject(s)
Bacterial Proteins/immunology , Mycobacterium tuberculosis/immunology , Th1 Cells/microbiology , Th17 Cells/microbiology , Tuberculosis/immunology , Virulence Factors/immunology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/physiology , Host-Pathogen Interactions , Interleukin-10/immunology , Interleukin-12/immunology , Lung/immunology , Lung/microbiology , Lung/pathology , Macrophages/immunology , Macrophages/microbiology , Macrophages/pathology , Mice, Inbred BALB C , Mice, Inbred C57BL , Mutation , Mycobacterium tuberculosis/genetics , Th1 Cells/immunology , Th1 Cells/pathology , Th17 Cells/immunology , Th17 Cells/pathology , Tuberculosis/pathology , Virulence Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...