Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
1.
Am J Respir Cell Mol Biol ; 59(5): 548-556, 2018 11.
Article in English | MEDLINE | ID: mdl-29852080

ABSTRACT

Vitamin A deficiency strongly predicts the risk of developing tuberculosis (TB) in individuals exposed to Mycobacterium tuberculosis (Mtb). The burden of antibiotic-resistant TB is increasing globally; therefore, there is an urgent need to develop host-directed adjunctive therapies to treat TB. Alveolar macrophages, the niche cell for Mtb, metabolize vitamin A to all-trans retinoic acid (atRA), which influences host immune responses. We sought to determine the mechanistic effects of atRA on the host immune response to intracellular bacterial infection in primary human and murine macrophages. In this study, atRA promoted autophagy resulting in a reduced bacterial burden in human macrophages infected with Mtb and Bordetella pertussis, but not bacillus Calmette-Guérin (BCG). Autophagy is induced by cytosolic sensing of double-stranded DNA via the STING/TBK1/IRF3 axis; however, BCG is known to evade cytosolic DNA sensors. atRA enhanced colocalization of Mtb, but not BCG, with autophagic vesicles and acidified lysosomes. This enhancement was inhibited by blocking TBK1. Our data indicate that atRA augments the autophagy of intracellular bacteria that trigger cytosolic DNA-sensing pathways but does not affect bacteria that evade these sensors. The finding that BCG evades the beneficial effects of atRA has implications for vaccine design and global health nutritional supplementation strategies. The ability of atRA to promote autophagy and aid bacterial clearance of Mtb and B. pertussis highlights a potential role for atRA as a host-directed adjunctive therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Antitubercular Agents/pharmacology , Autophagy , Macrophages, Alveolar/pathology , Mycobacterium tuberculosis/drug effects , Tretinoin/pharmacology , Tuberculosis/pathology , Cells, Cultured , Humans , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/microbiology , Tuberculosis/drug therapy , Tuberculosis/microbiology
2.
J Immunol ; 189(8): 4144-53, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22972933

ABSTRACT

Autophagy controls IL-1ß secretion by regulating inflammasome activation and by targeting pro-IL-1ß for degradation. In this article, we show that inhibition of autophagy, either with the PI3K inhibitors 3-methyladenine, wortmannin, and LY294002 or with small interfering RNA against autophagy proteins augmented the secretion of IL-23 by human and mouse macrophages and dendritic cells in response to specific TLR agonists. This process occurred at the transcriptional level and was dependent on reactive oxygen species and IL-1R signaling; it was abrogated with an IL-1R antagonist or with IL-1-neutralizing Abs, whereas treatment with either rIL-1α or IL-1ß induced IL-23 secretion. Dendritic cells treated with LPS and 3-methyladenine secreted enhanced levels of both IL-1ß and IL-23, and supernatants from these cells stimulated the innate secretion of IL-17, IFN-γ, and IL-22 by γδ T cells. These data demonstrate that autophagy has a potentially pivotal role to play in the induction and regulation of inflammatory responses by innate immune cells, largely driven by IL-1 and its consequential effects on IL-23 secretion.


Subject(s)
Autophagy/immunology , Interleukin-1alpha/metabolism , Interleukin-1beta/metabolism , Interleukin-23/metabolism , T-Lymphocyte Subsets/immunology , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Cell Line , Cell Line, Transformed , Cells, Cultured , Female , Immunity, Innate , Inflammation Mediators/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocyte Subsets/metabolism , Up-Regulation/immunology
3.
Autophagy ; 12(6): 907-16, 2016 06 02.
Article in English | MEDLINE | ID: mdl-27163877

ABSTRACT

MIF (macrophage migration inhibitory factor [glycosylation-inhibiting factor]) is a pro-inflammatory cytokine expressed in multiple cells types, including macrophages. MIF plays a pathogenic role in a number of inflammatory diseases and has been linked to tumor progression in some cancers. Previous work has demonstrated that loss of autophagy in macrophages enhances secretion of IL1 family cytokines. Here, we demonstrate that loss of autophagy, by pharmacological inhibition or siRNA silencing of Atg5, enhances MIF secretion by monocytes and macrophages. We further demonstrate that this is dependent on mitochondrial reactive oxygen species (ROS). Induction of autophagy with MTOR inhibitors had no effect on MIF secretion, but amino acid starvation increased secretion. This was unaffected by Atg5 siRNA but was again dependent on mitochondrial ROS. Our data demonstrate that autophagic regulation of mitochondrial ROS plays a pivotal role in the regulation of inflammatory cytokine secretion in macrophages, with potential implications for the pathogenesis of inflammatory diseases and cancers.


Subject(s)
Autophagy , Macrophage Migration-Inhibitory Factors/metabolism , Macrophages/metabolism , Amino Acids/deficiency , Animals , Humans , Mice , Mitochondria/metabolism , RAW 264.7 Cells , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL