Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Microbiol Spectr ; 12(1): e0498122, 2024 Jan 11.
Article in English | MEDLINE | ID: mdl-38051049

ABSTRACT

IMPORTANCE: Activation of the host transcription factor TFEB helps mammalian cells adapt to stresses such as starvation and infection by upregulating lysosome, autophagy, and immuno-protective gene expression. Thus, TFEB is generally thought to protect host cells. However, it may also be that pathogenic bacteria like Salmonella orchestrate TFEB in a spatio-temporal manner to harness its functions to grow intracellularly. Indeed, the relationship between Salmonella and TFEB is controversial since some studies showed that Salmonella actively promotes TFEB, while others have observed that Salmonella degrades TFEB and that compounds that promote TFEB restrict bacterial growth. Our work provides a path to resolve these apparent discordant observations since we showed that stationary-grown Salmonella actively delays TFEB after infection, while late-log Salmonella is permissive of TFEB activation. Nevertheless, the exact function of this manipulation remains unclear, but conditions that erase the conditional control of TFEB by Salmonella may be detrimental to the microbe.


Subject(s)
Macrophages , TOR Serine-Threonine Kinases , Animals , Mice , TOR Serine-Threonine Kinases/metabolism , Macrophages/metabolism , Autophagy/physiology , Lysosomes/physiology , Salmonella , Mammals
2.
Cell Microbiol ; 20(4)2018 04.
Article in English | MEDLINE | ID: mdl-29349904

ABSTRACT

Lysosomes are acidic and hydrolytic organelles responsible for receiving and digesting cargo acquired during endocytosis, phagocytosis, and autophagy. For macrophages and dendritic cells, the lysosome is kingpin, playing a direct role in microbe killing and antigen processing for presentation. Strikingly, the historic view that lysosomes are homogeneous and static organelles is being replaced with a more elegant paradigm, in which lysosomes are heterogeneous, dynamic, and respond to cellular needs. For example, lysosomes are signalling platforms that integrate stress detection and molecular decision hubs such as the mTOR complex 1 and AMPK to modulate cellular activity. These signals can even adjust lysosome activity by modulating transcription factors such as transcription factor EB (TFEB) and TFE3 that govern lysosome gene expression. Here, we review lysosome remodelling and adaptation during macrophage and dendritic cell stimulation. First, we assess the functional outcomes and regulatory mechanisms driving the dramatic restructuring of lysosomes from globular organelles into a tubular network during phagocyte activation. Second, we discuss lysosome adaptation and scaling in macrophages driven by TFEB and TFE3 stimulation in response to phagocytosis and microbe challenges. Collectively, we are beginning to appreciate that lysosomes are dynamic and adapt to serve phagocyte differentiation in response to microbes and immune stress.


Subject(s)
Lysosomes/physiology , Phagocytosis/physiology , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/physiology , Dendritic Cells/physiology , Humans , Macrophage Activation/physiology , Macrophages/physiology
3.
Curr Biol ; 26(15): 1955-1964, 2016 08 08.
Article in English | MEDLINE | ID: mdl-27397893

ABSTRACT

Macrophages internalize pathogens through phagocytosis, entrapping them into organelles called phagosomes. Phagosomes then fuse with lysosomes to mature into phagolysosomes, acquiring an acidic and hydrolytic lumen that kills the pathogens. During an ongoing infection, macrophages can internalize dozens of bacteria. Thus, we hypothesized that an initial round of phagocytosis might boost lysosome function and bactericidal ability to cope with subsequent rounds of phagocytosis. To test this hypothesis, we employed Fcγ-receptor-mediated phagocytosis and endocytosis, which internalize immunoglobulin G (IgG)-opsonized particles and polyvalent IgG immune complexes, respectively. We report that Fcγ receptor activation in macrophages enhances lysosome-based proteolysis and killing of subsequently phagocytosed E. coli compared to naive macrophages. Importantly, we show that Fcγ receptor activation causes nuclear translocation of TFEB, a transcription factor that boosts expression of lysosome genes. Indeed, Fc receptor activation is accompanied by increased expression of specific lysosomal proteins. Remarkably, TFEB silencing represses the Fcγ-receptor-mediated enhancements in degradation and bacterial killing. In addition, nuclear translocation of TFEB requires phagosome completion and fails to occur in cells silenced for MCOLN1, a lysosomal Ca(2+) channel, suggesting that lysosomal Ca(2+) released during phagosome maturation activates TFEB. Finally, we demonstrate that non-opsonic phagocytosis of E. coli also enhances lysosomal degradation in a TFEB-dependent manner, suggesting that this phenomenon is not limited to Fcγ receptors. Overall, we show that macrophages become better killers after one round of phagocytosis and suggest that phagosomes and lysosomes are capable of bi-directional signaling.


Subject(s)
Anti-Bacterial Agents/pharmacology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Lysosomes/physiology , Phagocytosis/physiology , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/immunology , Endocytosis , Escherichia coli/immunology , Mice , Protein Transport , RAW 264.7 Cells , Receptors, IgG/genetics , Receptors, IgG/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...