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.
PLoS Pathog ; 17(9): e1009943, 2021 09.
Article in English | MEDLINE | ID: mdl-34555129

ABSTRACT

Regulation of cellular metabolism is now recognized as a crucial mechanism for the activation of innate and adaptive immune cells upon diverse extracellular stimuli. Macrophages, for instance, increase glycolysis upon stimulation with pathogen-associated molecular patterns (PAMPs). Conceivably, pathogens also counteract these metabolic changes for their own survival in the host. Despite this dynamic interplay in host-pathogen interactions, the role of immunometabolism in the context of intracellular bacterial infections is still unclear. Here, employing unbiased metabolomic and transcriptomic approaches, we investigated the role of metabolic adaptations of macrophages upon Salmonella enterica serovar Typhimurium (S. Typhimurium) infections. Importantly, our results suggest that S. Typhimurium abrogates glycolysis and its modulators such as insulin-signaling to impair macrophage defense. Mechanistically, glycolysis facilitates glycolytic enzyme aldolase A mediated v-ATPase assembly and the acidification of phagosomes which is critical for lysosomal degradation. Thus, impairment in the glycolytic machinery eventually leads to decreased bacterial clearance and antigen presentation in murine macrophages (BMDM). Collectively, our results highlight a vital molecular link between metabolic adaptation and phagosome maturation in macrophages, which is targeted by S. Typhimurium to evade cell-autonomous defense.


Subject(s)
Glycolysis/physiology , Host-Pathogen Interactions/physiology , Macrophages/metabolism , Phagosomes/metabolism , Salmonella Infections, Animal/metabolism , Animals , Gene Expression Profiling , Metabolomics , Mice , Salmonella typhimurium/metabolism
2.
J Immunol ; 205(9): 2456-2467, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32948684

ABSTRACT

Salmonella enterica serovar Typhimurium (S Typhimurium) is a Gram-negative bacterium that induces cell death of macrophages as a key virulence strategy. We have previously demonstrated that the induction of macrophage death is dependent on the host's type I IFN (IFN-I) response. IFN-I signaling has been shown to induce tripartite motif (TRIM) 21, an E3 ubiquitin ligase with critical functions in autoimmune disease and antiviral immunity. However, the importance and regulation of TRIM21 during bacterial infection remains poorly understood. In this study, we investigated the role of TRIM21 upon S Typhimurium infection of murine bone marrow-derived macrophages. Although Trim21 expression was induced in an IFN-I-dependent manner, we found that TRIM21 levels were mainly regulated posttranscriptionally. Following TLR4 activation, TRIM21 was transiently degraded via the lysosomal pathway by chaperone-mediated autophagy (CMA). However, S Typhimurium-induced mTORC2 signaling led to phosphorylation of Akt at S473, which subsequently impaired TRIM21 degradation by attenuating CMA. Elevated TRIM21 levels promoted macrophage death associated with reduced transcription of NF erythroid 2-related factor 2 (NRF2)-dependent antioxidative genes. Collectively, our results identify IFN-I-inducible TRIM21 as a negative regulator of innate immune responses to S Typhimurium and a previously unrecognized substrate of CMA. To our knowledge, this is the first study reporting that a member of the TRIM family is degraded by the lysosomal pathway.


Subject(s)
Chaperone-Mediated Autophagy/immunology , Ribonucleoproteins/immunology , Ribonucleoproteins/metabolism , Salmonella Infections/immunology , Salmonella Infections/metabolism , Salmonella typhimurium/immunology , Animals , Immunity, Innate/immunology , Lysosomes/immunology , Lysosomes/metabolism , Macrophages/immunology , Macrophages/metabolism , Mechanistic Target of Rapamycin Complex 2/immunology , Mechanistic Target of Rapamycin Complex 2/metabolism , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/immunology , NF-E2-Related Factor 2/metabolism , Phosphorylation/immunology , Proto-Oncogene Proteins c-akt/immunology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/immunology
3.
Lancet Microbe ; 1(3): e119-e129, 2020 Jul.
Article in English | MEDLINE | ID: mdl-35544262

ABSTRACT

BACKGROUND: Sick newborns admitted to neonatal units in low-resource settings are at an increased risk of developing hospital-acquired infections due to poor clinical care practices. Clusters of infection, due to the same species, with a consistent antibiotic resistance profile, and in the same ward over a short period of time might be indicative of an outbreak. We used whole-genome sequencing (WGS) to define the transmission pathways and characterise two distinct outbreaks of neonatal bacteraemia in a west African neonatal unit. METHODS: We studied two outbreaks of Burkholderia cepacia and multidrug-resistant extended spectrum ß-lactamase (ESBL)-producing Klebsiella pneumoniae in a neonatal unit that provides non-intensive care on the neonatal ward in the Edward Francis Small Teaching Hospital, Banjul, The Gambia. We used WGS to validate and expand findings from the outbreak investigation. We retrospectively sequenced all clinical isolates associated with each outbreak, including isolates obtained from swabs of ward surfaces, environmental fluid cultures, intravenous fluids, and antibiotics administered to newborns. We also sequenced historical B cepacia isolates associated with neonatal sepsis in the same ward. RESULTS: Between March 1 and Dec 31, 2016, 321 blood cultures were done, of which 178 (55%) were positive with a clinically significant isolate. 49 episodes of neonatal B cepacia bacteraemia and 45 episodes of bacteraemia due to ESBL-producing K pneumoniae were reported. WGS revealed the suspected K pneumoniae outbreak to be contemporaneous outbreaks of K pneumoniae (ST39) and previously unreported Klebsiella quasipneumoniae subspecies similipneumoniae (ST1535). Genomic analysis showed near-identical strain clusters for each of the three outbreak pathogens, consistent with transmission within the neonatal ward from extrinsically contaminated in-use intravenous fluids and antibiotics. Time-dated phylogeny, including retrospective analysis of archived bacterial strains, suggest B cepacia has been endemic in the neonatal ward over several years, with the Klebsiella species a more recent introduction. INTERPRETATION: Our study highlights the emerging threat of previously unreported strains of multidrug-resistant Klebsiella species in this neonatal unit. Genome-based surveillance studies can improve identification of circulating pathogen strains, characterisation of antimicrobial resistance, and help understand probable infection acquisition routes during outbreaks in newborn units in low-resource settings. Our data provide evidence for the need to regularly monitor endemic transmission of bacteria within the hospital setting, identify the introduction of resistant strains from the community, and improve clinical practices to reduce or prevent the spread of infection and resistance. FUNDING: Medical Research Council Unit The Gambia at the London School of Hygiene & Tropical Medicine, Fajara, The Gambia.

4.
Proc Natl Acad Sci U S A ; 116(33): 16551-16560, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31350351

ABSTRACT

The dynamic interplay between metabolism and immune responses in health and disease, by which different immune cells impact on metabolic processes, are being increasingly appreciated. However, the potential of master regulators of metabolism to control innate immunity are less understood. Here, we studied the cross-talk between leptin signaling and macrophage function in the context of bacterial infections. We found that upon infection with Gram-negative pathogens, such as Salmonella Typhimurium, leptin receptor (Lepr) expression increased in both mouse and human macrophages. Unexpectedly, both genetic Lepr ablation in macrophages and global pharmacologic leptin antagonization augmented lysosomal functions, reduced S Typhimurium burden, and diminished inflammation in vitro and in vivo. Mechanistically, we show that leptin induction activates the mTORC2/Akt pathway and subsequently down-regulates Phlpp1 phosphatase, allowing for phosphorylated Akt to impair lysosomal-mediated pathogen clearance. These data highlight a link between leptin signaling, the mTORC2/Phlpp1/Akt axis, and lysosomal activity in macrophages and have important therapeutic implications for modulating innate immunity to combat Gram-negative bacterial infections.


Subject(s)
Leptin/metabolism , Macrophages/immunology , Salmonella typhimurium/immunology , Signal Transduction , Adult , Animals , Female , Humans , Inflammation/pathology , Leptin/antagonists & inhibitors , Lysosomes/metabolism , Macrophages/microbiology , Mechanistic Target of Rapamycin Complex 2/metabolism , Mice , Mice, Inbred C57BL , Models, Biological , Phagosomes/metabolism , Phosphoprotein Phosphatases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , RAW 264.7 Cells , Receptors, Leptin/metabolism , Salmonella Infections, Animal , Young Adult
5.
J Cell Biol ; 216(12): 4107-4121, 2017 12 04.
Article in English | MEDLINE | ID: mdl-29055012

ABSTRACT

Salmonella enterica serovar Typhimurium exploits the host's type I interferon (IFN-I) response to induce receptor-interacting protein (RIP) kinase-mediated necroptosis in macrophages. However, the events that drive necroptosis execution downstream of IFN-I and RIP signaling remain elusive. In this study, we demonstrate that S Typhimurium infection causes IFN-I-mediated up-regulation of the mitochondrial phosphatase Pgam5 through RIP3. Pgam5 subsequently interacts with Nrf2, which sequesters Nrf2 in the cytosol, thereby repressing the transcription of Nrf2-dependent antioxidative genes. The impaired ability to respond to S Typhimurium-induced oxidative stress results in reactive oxygen species-mediated mitochondrial damage, energy depletion, transient induction of autophagy, and autophagic degradation of p62. Reduced p62 levels impair interaction of p62 with Keap1, which further decreases Nrf2 function and antioxidative responses to S Typhimurium infection, eventually leading to cell death. Collectively, we identify impaired Nrf2-dependent redox homeostasis as an important mechanism that promotes cell death downstream of IFN-I and RIP3 signaling in S Typhimurium-infected macrophages.


Subject(s)
Apoptosis/genetics , Interferon Type I/immunology , Macrophages/immunology , NF-E2-Related Factor 2/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , Salmonella typhimurium/physiology , Animals , Autophagy/genetics , Bone Marrow Cells/immunology , Bone Marrow Cells/microbiology , Gene Expression Regulation , Interferon Type I/genetics , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/immunology , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/immunology , Mitochondria/microbiology , NF-E2-Related Factor 2/genetics , Necrosis/genetics , Necrosis/immunology , Necrosis/pathology , Oxidative Stress , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/immunology , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Sequestosome-1 Protein/genetics , Sequestosome-1 Protein/immunology
6.
PLoS Pathog ; 13(2): e1006227, 2017 02.
Article in English | MEDLINE | ID: mdl-28192515

ABSTRACT

During intracellular infections, autophagy significantly contributes to the elimination of pathogens, regulation of pro-inflammatory signaling, secretion of immune mediators and in coordinating the adaptive immune system. Intracellular pathogens such as S. Typhimurium have evolved mechanisms to circumvent autophagy. However, the regulatory mechanisms targeted by S. Typhimurium to modulate autophagy have not been fully resolved. Here we report that cytosolic energy loss during S. Typhimurium infection triggers transient activation of AMPK, an important checkpoint of mTOR activity and autophagy. The activation of AMPK is regulated by LKB1 in a cytosolic complex containing Sirt1 and LKB1, where Sirt1 is required for deacetylation and subsequent activation of LKB1. S. Typhimurium infection targets Sirt1, LKB1 and AMPK to lysosomes for rapid degradation resulting in the disruption of the AMPK-mediated regulation of mTOR and autophagy. The degradation of cytosolic Sirt1/LKB1/AMPK complex was not observed with two mutant strains of S. Typhimurium, ΔssrB and ΔssaV, both compromising the pathogenicity island 2 (SPI2). The results highlight virulence factor-dependent degradation of host cell proteins as a previously unrecognized strategy of S. Typhimurium to evade autophagy.


Subject(s)
AMP-Activated Protein Kinases/immunology , Autophagy/physiology , Salmonella Infections/immunology , Sirtuin 1/immunology , TOR Serine-Threonine Kinases/immunology , AMP-Activated Protein Kinases/metabolism , Animals , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Blotting, Western , Cell Cycle Checkpoints/physiology , Disease Models, Animal , Immunohistochemistry , Immunoprecipitation , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Protein Serine-Threonine Kinases/immunology , Protein Serine-Threonine Kinases/metabolism , Salmonella typhimurium/immunology , Salmonella typhimurium/pathogenicity , Signal Transduction/immunology , Sirtuin 1/metabolism , TOR Serine-Threonine Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...