Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
2.
Cell Host Microbe ; 27(2): 238-248.e7, 2020 02 12.
Article in English | MEDLINE | ID: mdl-31901518

ABSTRACT

Antimicrobial resistance in tuberculosis (TB) is a public health threat of global dimension, worsened by increasing drug resistance. Host-directed therapy (HDT) is an emerging concept currently explored as an adjunct therapeutic strategy for TB. One potential host target is the ligand-activated transcription factor aryl hydrocarbon receptor (AhR), which binds TB virulence factors and controls antibacterial responses. Here, we demonstrate that in the context of therapy, the AhR binds several TB drugs, including front line drugs rifampicin (RIF) and rifabutin (RFB), resulting in altered host defense and drug metabolism. AhR sensing of TB drugs modulates host defense mechanisms, notably impairs phagocytosis, and increases TB drug metabolism. Targeting AhR in vivo with a small-molecule inhibitor increases RFB-treatment efficacy. Thus, the AhR markedly impacts TB outcome by affecting both host defense and drug metabolism. As a corollary, we propose the AhR as a potential target for HDT in TB in adjunct to canonical chemotherapy.


Subject(s)
Antitubercular Agents/metabolism , Mycobacterium tuberculosis , Receptors, Aryl Hydrocarbon/drug effects , Tuberculosis/drug therapy , Animals , Antitubercular Agents/therapeutic use , Basic Helix-Loop-Helix Transcription Factors/drug effects , Basic Helix-Loop-Helix Transcription Factors/metabolism , Humans , Immunity, Cellular/drug effects , Mycobacterium marinum/drug effects , Mycobacterium marinum/pathogenicity , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/pathogenicity , Phagocytosis/drug effects , Receptors, Aryl Hydrocarbon/metabolism , Rifabutin/metabolism , Rifabutin/therapeutic use , Rifampin/metabolism , Rifampin/therapeutic use , THP-1 Cells , Treatment Outcome , Tuberculosis/microbiology , Zebrafish
3.
Science ; 366(6472)2019 12 20.
Article in English | MEDLINE | ID: mdl-31857448

ABSTRACT

Pseudomonas aeruginosa rapidly adapts to altered conditions by quorum sensing (QS), a communication system that it uses to collectively modify its behavior through the production, release, and detection of signaling molecules. QS molecules can also be sensed by hosts, although the respective receptors and signaling pathways are poorly understood. We describe a pattern of regulation in the host by the aryl hydrocarbon receptor (AhR) that is critically dependent on qualitative and quantitative sensing of P. aeruginosa quorum. QS molecules bind to AhR and distinctly modulate its activity. This is mirrored upon infection with P. aeruginosa collected from diverse growth stages and with QS mutants. We propose that by spying on bacterial quorum, AhR acts as a major sensor of infection dynamics, capable of orchestrating host defense according to the status quo of infection.


Subject(s)
Host-Pathogen Interactions , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/pathogenicity , Quorum Sensing/physiology , Receptors, Aryl Hydrocarbon/physiology , A549 Cells , Animals , Humans , Larva , Macrophages/microbiology , Mice , Mice, Knockout , Pseudomonas aeruginosa/genetics , Quorum Sensing/genetics , Receptors, Aryl Hydrocarbon/genetics , Zebrafish
4.
Sci Rep ; 9(1): 10878, 2019 07 26.
Article in English | MEDLINE | ID: mdl-31350436

ABSTRACT

As a first host barrier, the skin is constantly exposed to environmental insults that perturb its integrity. Tight regulation of skin homeostasis is largely controlled by the aryl hydrocarbon receptor (AhR). Here, we demonstrate that Henna and its major pigment, the naphthoquinone Lawsone activate AhR, both in vitro and in vivo. In human keratinocytes and epidermis equivalents, Lawsone exposure enhances the production of late epidermal proteins, impacts keratinocyte differentiation and proliferation, and regulates skin inflammation. To determine the potential use of Lawsone for therapeutic application, we harnessed human, murine and zebrafish models. In skin regeneration models, Lawsone interferes with physiological tissue regeneration and inhibits wound healing. Conversely, in a human acute dermatitis model, topical application of a Lawsone-containing cream ameliorates skin irritation. Altogether, our study reveals how a widely used natural plant pigment is sensed by the host receptor AhR, and how the physiopathological context determines beneficial and detrimental outcomes.


Subject(s)
Dermatitis/drug therapy , Keratinocytes/metabolism , Naphthoquinones/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Skin/metabolism , Animals , Cells, Cultured , Guided Tissue Regeneration , Homeostasis , Humans , Lawsonia Plant , Mice , Models, Animal , Naphthoquinones/therapeutic use , Skin/drug effects , Skin/pathology , Wound Healing , Zebrafish
5.
EMBO Rep ; 20(4)2019 04.
Article in English | MEDLINE | ID: mdl-30872316

ABSTRACT

Cyclic dinucleotides (CDNs) are important second messenger molecules in prokaryotes and eukaryotes. Within host cells, cytosolic CDNs are detected by STING and alert the host by activating innate immunity characterized by type I interferon (IFN) responses. Extracellular bacteria and dying cells can release CDNs, but sensing of extracellular CDNs (eCDNs) by mammalian cells remains elusive. Here, we report that endocytosis facilitates internalization of eCDNs. The DNA sensor cGAS facilitates sensing of endocytosed CDNs, their perinuclear accumulation, and subsequent STING-dependent release of type I IFN Internalized CDNs bind cGAS directly, leading to its dimerization, and the formation of a cGAS/STING complex, which may activate downstream signaling. Thus, eCDNs comprise microbe- and danger-associated molecular patterns that contribute to host-microbe crosstalk during health and disease.


Subject(s)
Host-Pathogen Interactions , Immunity, Innate , Nucleotides, Cyclic/metabolism , Nucleotidyltransferases/metabolism , Animals , Cell Line , Endocytosis/genetics , Endocytosis/immunology , Extracellular Space , Host-Pathogen Interactions/immunology , Humans , Interferon Type I/metabolism , Macrophages/immunology , Macrophages/metabolism , Membrane Proteins/metabolism , Mice , Models, Molecular , Nucleotides, Cyclic/chemistry , Nucleotidyltransferases/chemistry , Nucleotidyltransferases/genetics , Protein Binding , Protein Conformation , Protein Multimerization , Second Messenger Systems , Signal Transduction , Structure-Activity Relationship
6.
Sci Rep ; 8(1): 1520, 2018 01 24.
Article in English | MEDLINE | ID: mdl-29367626

ABSTRACT

Mycobacterium tuberculosis (Mtb) is a life-threatening pathogen in humans. Bacterial infection of macrophages usually triggers strong innate immune mechanisms, including IL-1 cytokine secretion. The newer member of the IL-1 family, IL-36, was recently shown to be involved in cellular defense against Mtb. To unveil the underlying mechanism of IL-36 induced antibacterial activity, we analyzed its role in the regulation of cholesterol metabolism, together with the involvement of Liver X Receptor (LXR) in this process. We report that, in Mtb-infected macrophages, IL-36 signaling modulates cholesterol biosynthesis and efflux via LXR. Moreover, IL-36 induces the expression of cholesterol-converting enzymes and the accumulation of LXR ligands, such as oxysterols. Ultimately, both IL-36 and LXR signaling play a role in the regulation of antimicrobial peptides expression and in Mtb growth restriction. These data provide novel evidence for the importance of IL-36 and cholesterol metabolism mediated by LXR in cellular host defense against Mtb.


Subject(s)
Cholesterol/metabolism , Host-Pathogen Interactions , Interleukin-1/metabolism , Liver X Receptors/metabolism , Mycobacterium tuberculosis/immunology , Tuberculosis/immunology , Antimicrobial Cationic Peptides/biosynthesis , HEK293 Cells , Humans , Macrophages/immunology , Macrophages/microbiology , THP-1 Cells
7.
J Infect Dis ; 214(3): 464-74, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27389350

ABSTRACT

Tuberculosis remains a major killer worldwide, not the least because of our incomplete knowledge of protective and pathogenic immune mechanism. The roles of the interleukin 1 (IL-1) and interleukin 18 pathways in host defense are well established, as are their regulation through the inflammasome complex. In contrast, the regulation of interleukin 36γ (IL-36γ), a recently described member of the IL-1 family, and its immunological relevance in host defense remain largely unknown. Here we show that Mycobacterium tuberculosis infection of macrophages induces IL-36γ production in a 2-stage-regulated fashion. In the first stage, microbial ligands trigger host Toll-like receptor and MyD88-dependent pathways, leading to IL-36γ secretion. In the second stage, endogenous IL-1ß and interleukin 18 further amplify IL-36γ synthesis. The relevance of this cytokine in the control of M. tuberculosis is demonstrated by IL-36γ-induced antimicrobial peptides and IL-36 receptor-dependent restriction of M. tuberculosis growth. Thus, we provide first insight into the induction and regulation of the proinflammatory cytokine IL-36γ during tuberculosis.


Subject(s)
Interleukin-1/metabolism , Mycobacterium tuberculosis/immunology , Tuberculosis/immunology , Animals , Cell Line , Humans , Interleukin-1/deficiency , Macrophages/immunology , Macrophages/microbiology , Mice, Inbred C57BL , Mice, Knockout
8.
Nature ; 512(7515): 387-92, 2014 Aug 28.
Article in English | MEDLINE | ID: mdl-25119038

ABSTRACT

The aryl hydrocarbon receptor (AhR) is a highly conserved ligand-dependent transcription factor that senses environmental toxins and endogenous ligands, thereby inducing detoxifying enzymes and modulating immune cell differentiation and responses. We hypothesized that AhR evolved to sense not only environmental pollutants but also microbial insults. We characterized bacterial pigmented virulence factors, namely the phenazines from Pseudomonas aeruginosa and the naphthoquinone phthiocol from Mycobacterium tuberculosis, as ligands of AhR. Upon ligand binding, AhR activation leads to virulence factor degradation and regulated cytokine and chemokine production. The relevance of AhR to host defence is underlined by heightened susceptibility of AhR-deficient mice to both P. aeruginosa and M. tuberculosis. Thus, we demonstrate that AhR senses distinct bacterial virulence factors and controls antibacterial responses, supporting a previously unidentified role for AhR as an intracellular pattern recognition receptor, and identify bacterial pigments as a new class of pathogen-associated molecular patterns.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Mycobacterium tuberculosis/immunology , Pigments, Biological/metabolism , Pseudomonas aeruginosa/immunology , Receptors, Aryl Hydrocarbon/metabolism , Receptors, Pattern Recognition/metabolism , Animals , Anti-Bacterial Agents/metabolism , Bone Marrow Cells/cytology , Cytokines/immunology , Cytokines/metabolism , Feedback, Physiological , Humans , Ligands , Macrophage Activation , Mice , Mycobacterium tuberculosis/growth & development , Mycobacterium tuberculosis/metabolism , Phenazines/metabolism , Pigments, Biological/chemistry , Pseudomonas Infections/metabolism , Pseudomonas aeruginosa/metabolism , Pyocyanine/metabolism , Virulence Factors/chemistry , Virulence Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...