Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Neurogastroenterol Motil ; : e14850, 2024 Jun 24.
Article in English | MEDLINE | ID: mdl-38924329

ABSTRACT

BACKGROUND: The natural history and pathophysiology of diverticular disease (DD) are still uncertain. An ex-vivo human complicated DD (cDD) model has recently shown a predominant transmural oxidative imbalance. The present study aims to evaluate whether the previously described alterations may precede the symptomatic form of the disease. METHODS: Colonic surgical samples obtained from patients with asymptomatic diverticulosis (DIV), complicated DD, and controls were systematically and detailed morphologically and molecularly analyzed. Therefore, histologic, histomorphometric, immunohistochemical evaluation, and gene and protein expression analysis were performed to characterize colonic muscle changes and evaluate chronic inflammation, oxidative imbalance, and hypoxia. Functional muscle activity was tested on strips and isolated cells in response to contractile and relaxant agents. KEY RESULTS: Compared with controls, DD showed a marketed increase in muscle layer thickness, smooth muscle cell syncytium disarray, and increased interstitial fibrosis; moreover, the observed features were more evident in the cDD group. These changes mainly affected longitudinal muscle and were associated with altered contraction-relaxation dynamics and fibrogenic switch of smooth muscle cells. Chronic lymphoplasmacytic inflammation was primarily evident in the mucosa and spared the muscle. A transmural increase in carbonylated and nitrated proteins, with loss of antioxidant molecules, characterized both stages of DD, suggesting early oxidative stress probably triggered by recurrent ischemic events, more pronounced in cDD, where HIF-1 was detected in both muscle and mucosa. CONCLUSION & INFERENCES: The different DD clinical scenarios are part of a progressive process, with oxidative imbalance representing a new target in the management of DD.

2.
Elife ; 112022 09 20.
Article in English | MEDLINE | ID: mdl-36125130

ABSTRACT

During obesity and high fat-diet (HFD) feeding in mice, sustained low-grade inflammation includes not only increased pro-inflammatory macrophages in the expanding adipose tissue, but also bone marrow (BM) production of invasive Ly6Chigh monocytes. As BM adiposity also accrues with HFD, we explored the relationship between the gains in BM white adipocytes and invasive Ly6Chigh monocytes by in vivo and ex vivo paradigms. We find a temporal and causal link between BM adipocyte whitening and the Ly6Chigh monocyte surge, preceding the adipose tissue macrophage rise during HFD in mice. Phenocopying this, ex vivo treatment of BM cells with conditioned media from BM adipocytes or bona fide white adipocytes favoured Ly6Chigh monocyte preponderance. Notably, Ly6Chigh skewing was preceded by monocyte metabolic reprogramming towards glycolysis, reduced oxidative potential and increased mitochondrial fission. In sum, short-term HFD changes BM cellularity, resulting in local adipocyte whitening driving a gradual increase and activation of invasive Ly6Chigh monocytes.


Subject(s)
Bone Marrow , Monocytes , Adipocytes , Animals , Culture Media, Conditioned , Mice , Mice, Inbred C57BL , Monocytes/metabolism , Obesity/metabolism
3.
Nat Neurosci ; 24(7): 930-940, 2021 07.
Article in English | MEDLINE | ID: mdl-33795885

ABSTRACT

The neurodegenerative disease spinal muscular atrophy (SMA) is caused by deficiency in the survival motor neuron (SMN) protein. Currently approved SMA treatments aim to restore SMN, but the potential for SMN expression beyond physiological levels is a unique feature of adeno-associated virus serotype 9 (AAV9)-SMN gene therapy. Here, we show that long-term AAV9-mediated SMN overexpression in mouse models induces dose-dependent, late-onset motor dysfunction associated with loss of proprioceptive synapses and neurodegeneration. Mechanistically, aggregation of overexpressed SMN in the cytoplasm of motor circuit neurons sequesters components of small nuclear ribonucleoproteins, leading to splicing dysregulation and widespread transcriptome abnormalities with prominent signatures of neuroinflammation and the innate immune response. Thus, long-term SMN overexpression interferes with RNA regulation and triggers SMA-like pathogenic events through toxic gain-of-function mechanisms. These unanticipated, SMN-dependent and neuron-specific liabilities warrant caution on the long-term safety of treating individuals with SMA with AAV9-SMN and the risks of uncontrolled protein expression by gene therapy.


Subject(s)
Motor Neurons/metabolism , Motor Neurons/pathology , Nerve Degeneration , Survival of Motor Neuron 1 Protein/toxicity , Animals , Dependovirus , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Gene Transfer Techniques , Genetic Therapy/adverse effects , Genetic Vectors , Injections, Intraventricular , Mice , Motor Disorders/genetics , Motor Disorders/metabolism , Motor Disorders/pathology , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Survival of Motor Neuron 1 Protein/genetics
4.
Cell Rep ; 34(4): 108677, 2021 01 26.
Article in English | MEDLINE | ID: mdl-33503439

ABSTRACT

Pioneering studies from the early 1980s suggested that bacterial peptidoglycan-derived muramyl peptides (MPs) could exert either stimulatory or immunosuppressive functions depending, in part, on chronicity of exposure. However, this Janus-faced property of MPs remains largely unexplored. Here, we demonstrate the immunosuppressive potential of Nod1, the bacterial sensor of diaminopimelic acid (DAP)-containing MPs. Using a model of self-limiting peritonitis, we show that systemic Nod1 activation promotes an autophagy-dependent reprogramming of macrophages toward an alternative phenotype. Moreover, Nod1 stimulation induces the expansion of myeloid-derived suppressor cells (MDSCs) and maintains their immunosuppressive potential via arginase-1 activity. Supporting the role of MDSCs and tumor-associated macrophages in cancer, we demonstrate that myeloid-intrinsic Nod1 expression sustains intra-tumoral arginase-1 levels to foster an immunosuppressive and tumor-permissive microenvironment during colorectal cancer (CRC) development. Our findings support the notion that bacterial products, via Nod1 detection, modulate the immunosuppressive activity of myeloid cells and fuel tumor progression in CRC.


Subject(s)
Colorectal Neoplasms/immunology , Myeloid-Derived Suppressor Cells/immunology , Nod1 Signaling Adaptor Protein/immunology , Animals , Carcinogenesis/immunology , Colorectal Neoplasms/pathology , Female , Humans , Male , Mice , Tumor Microenvironment/immunology
5.
Cell Host Microbe ; 23(5): 644-652.e5, 2018 05 09.
Article in English | MEDLINE | ID: mdl-29746835

ABSTRACT

In physiological settings, the complement protein C3 is deposited on all bacteria, including invasive pathogens. However, because experimental host-bacteria systems typically use decomplemented serum to avoid the lytic action of complement, the impact of C3 coating on epithelial cell responses to invasive bacteria remains unexplored. Here, we demonstrate that following invasion, intracellular C3-positive Listeria monocytogenes is targeted by autophagy through a direct C3/ATG16L1 interaction, resulting in autophagy-dependent bacterial growth restriction. In contrast, Shigella flexneri and Salmonella Typhimurium escape autophagy-mediated growth restriction in part through the action of bacterial outer membrane proteases that cleave bound C3. Upon oral infection with Listeria, C3-deficient mice displayed defective clearance at the intestinal mucosa. Together, these results demonstrate an intracellular role of complement in triggering antibacterial autophagy and immunity against intracellular pathogens. Since C3 indiscriminately associates with foreign surfaces, the C3-ATG16L1 interaction may provide a universal mechanism of xenophagy initiation.


Subject(s)
Autophagy/drug effects , Autophagy/immunology , Bacteria/immunology , Carrier Proteins/immunology , Complement C3/immunology , Complement C3/pharmacology , Host-Pathogen Interactions/immunology , Animals , Autophagy-Related Proteins , Bacteria/pathogenicity , Bacterial Outer Membrane Proteins/immunology , Dysentery, Bacillary/immunology , Dysentery, Bacillary/microbiology , Epithelial Cells , Female , HCT116 Cells , HEK293 Cells , HeLa Cells , Humans , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Listeria monocytogenes/immunology , Listeria monocytogenes/pathogenicity , Listeriosis/immunology , Listeriosis/microbiology , Male , Mice , Mice, Inbred C57BL , Salmonella Infections/immunology , Salmonella Infections/microbiology , Salmonella typhimurium/immunology , Salmonella typhimurium/pathogenicity , Shigella flexneri/immunology , Shigella flexneri/pathogenicity , THP-1 Cells
6.
J Endocr Soc ; 2(4): 336-347, 2018 Apr 01.
Article in English | MEDLINE | ID: mdl-29577109

ABSTRACT

Nod-like receptor (NLR)X1 is an NLR family protein that localizes to the mitochondrial matrix and modulates reactive oxygen species production, possibly by directly interacting with the electron transport chain. Recent work demonstrated that cells lacking NLRX1 have higher oxygen consumption but lower levels of adenosine triphosphate, suggesting that NLRX1 might prevent uncoupling of oxidative phosphorylation. We therefore hypothesized that NLRX1 might regulate whole-body energy metabolism through its effect on mitochondria. Male NLRX1 whole-body knockout (KO) mice and wild-type (WT) C57BL/6N controls were fed a low-fat or a high-fat (HF) diet for 16 weeks from weaning. Contrary to this hypothesis, there were no differences in body weight, adiposity, energy intake, or energy expenditure between HF-fed KO and WT mice, but instead HF KO mice were partially protected from the development of diet-induced hyperglycemia. Additionally, HF KO mice did not present with hyperinsulinemia during the glucose tolerance test, as did HF WT mice. There were no genotype differences in insulin tolerance, which led us to consider a pancreatic phenotype. Histology revealed that KO mice were protected from HF-induced pancreatic lipid accumulation, suggesting a potential role for NLRX1 in pancreatic dysfunction during the development diet-induced type 2 diabetes mellitus. Hence, NLRX1 depletion partially protects against postabsorptive hyperglycemia in obesity that may be linked to the prevention of pancreatic lipid accumulation. Although the actual mechanisms restoring glucose and insulin dynamics remain unknown, NLRX1 emerges as a potentially interesting target to inhibit for the prevention of type 2 diabetes mellitus.

7.
Cell Rep ; 21(13): 3653-3661, 2017 12 26.
Article in English | MEDLINE | ID: mdl-29281815

ABSTRACT

NLRP6 is a Nod-like receptor expressed in the intestinal epithelium. Previous studies reported a protective role for NLRP6 against intestinal injury and colitis-associated carcinogenesis via the regulation and establishment of a healthy microbiota. However, these results were not obtained using littermate animals, leaving the possibility that the pro-colitogenic microbiota phenotype associated with knockout (KO) mice was stochastically acquired and genotype independent. Here, we analyzed the microbiota at three intestinal locations from Nlrp6-/- and wild-type (WT) littermates, either co-caged or individually caged after weaning. Our results demonstrate that NLRP6 does not significantly influence the intestinal microbiota at homeostasis, and they support a previously reported sex-biased microbial community structure. Moreover, WT and Nlrp6-/- littermate mice displayed comparable sensitivity to dextran sulfate sodium (DSS)-induced colitis, although increased sensitivity was noted in KO females. Our results clarify the role of NLRP6 in microbiota and colitis control, and they highlight the importance of analyzing littermate animals in such studies.


Subject(s)
Gastrointestinal Microbiome , Receptors, Cell Surface/metabolism , Animals , Colitis/chemically induced , Colitis/microbiology , Colitis/pathology , Dextran Sulfate , Female , Genotype , Male , Mice, Inbred C57BL , Mice, Knockout , Microbiota , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics
8.
BMC Genomics ; 17: 680, 2016 08 26.
Article in English | MEDLINE | ID: mdl-27561422

ABSTRACT

BACKGROUND: The intestinal epithelium plays a critical role in nutrient absorption and innate immune defense. Recent studies showed that metabolic stress pathways, in particular the integrated stress response (ISR), control intestinal epithelial cell fate and function. Here, we used RNA-seq to analyze the global transcript level and alternative splicing responses of primary murine enteroids undergoing two distinct ISR-triggering stresses, endoplasmic reticulum (ER) stress and nutrient starvation. RESULTS: Our results reveal the core transcript level response to ISR-associated stress in murine enteroids, which includes induction of stress transcription factors, as well as genes associated with chemotaxis and inflammation. We also identified the transcript expression signatures that are unique to each ISR stress. Among these, we observed that ER stress and nutrient starvation had opposite effects on intestinal stem cell (ISC) transcriptional reprogramming. In agreement, ER stress decreased EdU incorporation, a marker of cell proliferation, in primary murine enteroids, while nutrient starvation had an opposite effect. We also analyzed the impact of these cellular stresses on mRNA splicing regulation. Splicing events commonly regulated by both stresses affected genes regulating splicing and were associated with nonsense-mediated decay (NMD), suggesting that splicing is modulated by an auto-regulatory feedback loop during stress. In addition, we also identified a number of genes displaying stress-specific splicing regulation. We suggest that functional gene expression diversity may arise during stress by the coordination of alternative splicing and alternative translation, and that this diversity might contribute to the cellular response to stress. CONCLUSIONS: Together, these results provide novel understanding of the importance of metabolic stress pathways in the intestinal epithelium. Specifically, the importance of cellular stresses in the regulation of immune and defense function, metabolism, proliferation and ISC activity in the intestinal epithelium is highlighted. Furthermore, this work highlights an under-appreciated role played by alternative splicing in shaping the response to stress and reveals a potential mechanism for gene regulation involving coupling of AS and alternative translation start sites.


Subject(s)
Endoplasmic Reticulum Stress/genetics , Intestinal Mucosa/metabolism , RNA Splicing , Starvation/genetics , Transcriptome , Animals , Computational Biology/methods , Gene Expression Profiling , Gene Expression Regulation , Introns , Mice , Organoids , Stem Cells/metabolism , Transcription Initiation Site
9.
Cell Rep ; 14(11): 2576-86, 2016 Mar 22.
Article in English | MEDLINE | ID: mdl-26971996

ABSTRACT

The mitochondrial Nod-like receptor protein NLRX1 protects against colorectal tumorigenesis through mechanisms that remain unclear. Using mice with an intestinal epithelial cells (IEC)-specific deletion of Nlrx1, we find that NLRX1 provides an IEC-intrinsic protection against colitis-associated carcinogenesis in the colon. These Nlrx1 mutant mice have increased expression of Tnf, Egf, and Tgfb1, three factors essential for wound healing, as well as increased epithelial proliferation during the epithelial regeneration phase following injury triggered by dextran sodium sulfate. In primary intestinal organoids lacking Nlrx1, stimulation with TNF resulted in exacerbated proliferation and expression of the intestinal stem cell markers Olfm4 and Myb. This hyper-proliferation response was associated with increased activation of Akt and NF-κB pathways in response to TNF stimulation. Together, these results identify NLRX1 as a suppressor of colonic tumorigenesis that acts by controlling epithelial proliferation in the intestine during the regeneration phase following mucosal injury.


Subject(s)
Mitochondrial Proteins/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Blotting, Western , Carcinogenesis , Cell Proliferation , Colon/pathology , Colonic Neoplasms/chemically induced , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Dextran Sulfate/toxicity , Female , Glycoproteins/genetics , Glycoproteins/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Ki-67 Antigen/genetics , Ki-67 Antigen/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Proto-Oncogene Proteins c-myb/genetics , Proto-Oncogene Proteins c-myb/metabolism , Real-Time Polymerase Chain Reaction , Survival Rate
10.
J Biol Chem ; 290(34): 20904-20918, 2015 Aug 21.
Article in English | MEDLINE | ID: mdl-26134566

ABSTRACT

Invasive bacterial pathogens induce an amino acid starvation (AAS) response in infected host cells that controls host defense in part by promoting autophagy. However, whether AAS has additional significant effects on the host response to intracellular bacteria remains poorly characterized. Here we showed that Shigella, Salmonella, and Listeria interfere with spliceosomal U snRNA maturation in the cytosol. Bacterial infection resulted in the rerouting of U snRNAs and their cytoplasmic escort, the survival motor neuron (SMN) complex, to processing bodies, thus forming U snRNA bodies (U bodies). This process likely contributes to the decline in the cytosolic levels of U snRNAs and of the SMN complex proteins SMN and DDX20 that we observed in infected cells. U body formation was triggered by membrane damage in infected cells and was associated with the induction of metabolic stresses, such as AAS or endoplasmic reticulum stress. Mechanistically, targeting of U snRNAs to U bodies was regulated by translation initiation inhibition and the ATF4/ATF3 pathway, and U bodies rapidly disappeared upon removal of the stress, suggesting that their accumulation represented an adaptive response to metabolic stress. Importantly, this process likely contributed to shape the host response to invasive bacteria because down-regulation of DDX20 expression using short hairpin RNA (shRNA) amplified ATF3- and NF-κB-dependent signaling. Together, these results identify a critical role for metabolic stress and invasive bacterial pathogens in U body formation and suggest that this process contributes to host defense.


Subject(s)
Host-Pathogen Interactions/genetics , Listeria monocytogenes/metabolism , RNA, Small Nuclear/metabolism , Salmonella typhimurium/metabolism , Shigella flexneri/metabolism , Stress, Physiological/genetics , Survival of Motor Neuron 1 Protein/metabolism , Activating Transcription Factor 3/genetics , Activating Transcription Factor 3/metabolism , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Cell Membrane/metabolism , Cytoplasm/metabolism , Cytoplasm/microbiology , DEAD Box Protein 20/antagonists & inhibitors , DEAD Box Protein 20/genetics , DEAD Box Protein 20/metabolism , Gene Expression Regulation , HeLa Cells , Humans , Listeria monocytogenes/pathogenicity , NF-kappa B/genetics , NF-kappa B/metabolism , Peptide Chain Initiation, Translational , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA, Small Nuclear/genetics , RNA, Small Nuclear/ultrastructure , Salmonella typhimurium/pathogenicity , Shigella flexneri/pathogenicity , Signal Transduction , Spliceosomes/metabolism , Spliceosomes/microbiology , Survival of Motor Neuron 1 Protein/genetics
11.
J Biol Chem ; 289(28): 19317-30, 2014 Jul 11.
Article in English | MEDLINE | ID: mdl-24867956

ABSTRACT

NLRX1 is a mitochondrial Nod-like receptor (NLR) protein whose function remains enigmatic. Here, we observed that NLRX1 expression was glucose-regulated and blunted by SV40 transformation. In transformed but not primary murine embryonic fibroblasts, NLRX1 expression mediated resistance to an extrinsic apoptotic signal, whereas conferring susceptibility to intrinsic apoptotic signals, such as glycolysis inhibition, increased cytosolic calcium and endoplasmic reticulum stress. In a murine model of colorectal cancer induced by azoxymethane, NLRX1-/- mice developed fewer tumors than wild type mice. In contrast, in a colitis-associated cancer model combining azoxymethane and dextran sulfate sodium, NLRX1-/- mice developed a more severe pathology likely due to the increased sensitivity to dextran sulfate sodium colitis. Together, these results identify NLRX1 as a critical mitochondrial protein implicated in the regulation of apoptosis in cancer cells. The unique capacity of NLRX1 to regulate the cellular sensitivity toward intrinsic versus extrinsic apoptotic signals suggests a critical role for this protein in numerous physiological processes and pathological conditions.


Subject(s)
Apoptosis , Colitis/metabolism , Colonic Neoplasms/metabolism , Mitochondrial Proteins/metabolism , Neoplasm Proteins/metabolism , Animals , Cell Line, Transformed , Colitis/chemically induced , Colitis/genetics , Colitis/pathology , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Dextran Sulfate/toxicity , Mice , Mice, Knockout , Mitochondrial Proteins/genetics , Neoplasm Proteins/genetics
12.
Proc Natl Acad Sci U S A ; 111(20): E2110-9, 2014 May 20.
Article in English | MEDLINE | ID: mdl-24799673

ABSTRACT

To subvert host immunity, influenza A virus (IAV) induces early apoptosis in innate immune cells by disrupting mitochondria membrane potential via its polymerase basic protein 1-frame 2 (PB1-F2) accessory protein. Whether immune cells have mechanisms to counteract PB1-F2-mediated apoptosis is currently unknown. Herein, we define that the host mitochondrial protein nucleotide-binding oligomerization domain-like receptor (NLR)X1 binds to viral protein PB1-F2, preventing IAV-induced macrophage apoptosis and promoting both macrophage survival and type I IFN signaling. We initially observed that Nlrx1-deficient mice infected with IAV exhibited increased pulmonary viral replication, as well as enhanced inflammatory-associated pulmonary dysfunction and morbidity. Analysis of the lungs of IAV-infected mice revealed markedly enhanced leukocyte recruitment but impaired production of type I IFN in Nlrx1(-/-) mice. Impaired type I IFN production and enhanced viral replication was recapitulated in Nlrx1(-/-) macrophages and was associated with increased mitochondrial mediated apoptosis. Through gain- and loss-of-function strategies for protein interaction, we identified that NLRX1 directly bound PB1-F2 in the mitochondria of macrophages. Using a recombinant virus lacking PB1-F2, we confirmed that deletion of PB1-F2 abrogated NLRX1-dependent macrophage type I IFN production and apoptosis. Thus, our results demonstrate that NLRX1 acts as a mitochondrial sentinel protecting macrophages from PB1-F2-induced apoptosis and preserving their antiviral function. We further propose that NLRX1 is critical for macrophage immunity against IAV infection by sensing the extent of viral replication and maintaining a protective balance between antiviral immunity and excessive inflammation within the lungs.


Subject(s)
Apoptosis , Influenza A virus/immunology , Macrophages/immunology , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Viral Proteins/metabolism , Animals , Cell Line, Tumor , Humans , Inflammation , Influenza A virus/physiology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondrial Proteins/metabolism , Protein Binding , Protein Structure, Tertiary , Virus Replication
13.
Microb Cell ; 1(1): 48-50, 2014 Jan 06.
Article in English | MEDLINE | ID: mdl-28357210

ABSTRACT

Listeria monocytogenes is a Gram-positive bacterial pathogen that induces its own uptake in non-phagocytic cells. Following invasion, Listeria escapes from the entry vacuole through the secretion of a pore-forming toxin, listeriolysin O (LLO) that acts to damage and disrupt the vacuole membrane. Listeria then replicates in the cytosol and is able to spread from cell-to-cell using actin-based motility. In addition to LLO, Listeria produces two phospholipase toxins, a phosphatidylinositol-specific phospholipase C (PI-PLC, encoded by plcB) and a broad-range phospholipase C (PC-PLC, encoded by plcA), which contribute to bacterial virulence. It has long been recognized that secretion of PI- and PC-PLC enables the disruption of the double membrane vacuole during cell-to-cell spread, and those phospholipases have also been shown to augment LLO-dependent escape from the entry endosome. However, a specific role for Listeria phospholipases during the cytosolic stage of infection has not been previously reported. In a recent study, we demonstrated that Listeria PI-PLC and PC-PLC contribute to the bacterial escape from autophagy through a mechanism that involves direct inhibition of the autophagic flux in the infected cells [Tattoli et al. EMBO J (2013), 32, 3066-3078].

14.
EMBO J ; 32(23): 3066-78, 2013 Nov 27.
Article in English | MEDLINE | ID: mdl-24162724

ABSTRACT

Listeria can escape host autophagy defense pathways through mechanisms that remain poorly understood. We show here that in epithelial cells, Listeriolysin (LLO)-dependent cytosolic escape of Listeria triggered a transient amino-acid starvation host response characterized by GCN2 phosphorylation, ATF3 induction and mTOR inhibition, the latter favouring a pro-autophagic cellular environment. Surprisingly, rapid recovery of mTOR signalling was neither sufficient nor necessary for Listeria avoidance of autophagic targeting. Instead, we observed that Listeria phospholipases PlcA and PlcB reduced autophagic flux and phosphatidylinositol 3-phosphate (PI3P) levels, causing pre-autophagosomal structure stalling and preventing efficient targeting of cytosolic bacteria. In co-infection experiments, wild-type Listeria protected PlcA/B-deficient bacteria from autophagy-mediated clearance. Thus, our results uncover a critical role for Listeria phospholipases C in the inhibition of autophagic flux, favouring bacterial escape from host autophagic defense.


Subject(s)
Autophagy , Listeria monocytogenes/enzymology , Listeriosis/pathology , Phagosomes/pathology , Phospholipases/metabolism , Activating Transcription Factor 3/genetics , Activating Transcription Factor 3/metabolism , Animals , Bacterial Toxins/pharmacology , Blotting, Western , Cell Proliferation , Cells, Cultured , Cytosol/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Embryo, Mammalian/microbiology , Enzyme-Linked Immunosorbent Assay , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/microbiology , Fluorescent Antibody Technique , HeLa Cells , Heat-Shock Proteins/pharmacology , Hemolysin Proteins/pharmacology , Humans , Listeriosis/metabolism , Listeriosis/microbiology , Mice , Phagosomes/metabolism , Phosphatidylinositol Phosphates/genetics , Phosphatidylinositol Phosphates/metabolism , Phospholipases/genetics , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
15.
EMBO Rep ; 14(10): 900-6, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24008845

ABSTRACT

The cyclic dinucleotides 3'-5'diadenylate (c-diAMP) and 3'-5' diguanylate (c-diGMP) are important bacterial second messengers that have recently been shown to stimulate the secretion of type I Interferons (IFN-Is) through the c-diGMP-binding protein MPYS/STING. Here, we show that physiologically relevant levels of cyclic dinucleotides also stimulate a robust secretion of IL-1ß through the NLRP3 inflammasome. Intriguingly, this response is independent of MPYS/STING. Consistent with most NLRP3 inflammasome activators, the response to c-diGMP is dependent on the mobilization of potassium and calcium ions. However, in contrast to other NLRP3 inflammasome activators, this response is not associated with significant changes in mitochondrial potential or the generation of mitochondrial reactive oxygen species. Thus, cyclic dinucleotides activate the NLRP3 inflammasome through a unique pathway that could have evolved to detect pervasive bacterial pathogen-associated molecular patterns associated with intracellular infections.


Subject(s)
Carrier Proteins/metabolism , Cyclic GMP/analogs & derivatives , Dinucleoside Phosphates/pharmacology , Inflammasomes/metabolism , Animals , Calcium/metabolism , Carrier Proteins/genetics , Cell Line, Tumor , Cyclic GMP/pharmacology , Humans , Interleukin-1beta/metabolism , Macrophages/drug effects , Macrophages/metabolism , Membrane Potential, Mitochondrial , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein , Potassium/metabolism , Reactive Oxygen Species/metabolism
16.
Innate Immun ; 19(4): 438-48, 2013.
Article in English | MEDLINE | ID: mdl-23212541

ABSTRACT

NLRX1 is a member of the Nod-like receptor family of intracellular sensors of microbial- and danger-associated molecular patterns. NLRX1 has a N-terminal mitochondrial addressing sequence that localizes the protein to the mitochondrial matrix. Recently, conflicting reports have been presented with regard to the putative implication of NLRX1 as a negative regulator of MAVS-dependent cytosolic antiviral responses. Here, we generated a new NLRX1 knockout mouse strain and observed that bone marrow-derived macrophages and murine embryonic fibroblasts from NLRX1-deficient mice displayed normal antiviral and inflammatory responses following Sendai virus infection. Importantly, wild type and NLRX1-deficient mice exhibited unaltered antiviral and inflammatory gene expression following intranasal challenge with influenza A virus or i.p. injection of Poly (I:C). Together, our results demonstrate that NLRX1 does not participate in the negative regulation of MAVS-dependent antiviral responses.


Subject(s)
Fibroblasts/immunology , Influenza A virus/immunology , Macrophages/immunology , Mitochondrial Proteins/metabolism , Orthomyxoviridae Infections/immunology , Respirovirus Infections/immunology , Sendai virus/immunology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Line , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , Immunity, Innate , Inflammation Mediators/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Proteins/genetics , Poly I-C/immunology , Signal Transduction
17.
Biol Open ; 1(12): 1215-25, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23259056

ABSTRACT

Bacterial invasion results in the rapid induction of an acute state of cytosolic amino acid (AA) starvation, provoked by host membrane damage. Bacteria-induced AA starvation, in turn, down-regulates mTOR signaling while triggering autophagy and the integrated stress response pathway dependent on GCN2, eIF2α and ATF3. In Salmonella-infected cells, we now demonstrate that the host AA starvation response program depended on the Salmonella pathogenicity island (SPI)-1, the activity of which was required to damage the Salmonella-containing vacuole (SCV) in the early stage of infection. At a later stage (3-4 hour post-infection), the progressive recruitment of mTOR to the surface of the SCV appeared to be independent of the activity of SPI-2 and of SCV positioning in the cell. Instead, mTOR localization to the SCV required the activity of host AA transporters SLC1A5, SLC3A2 and SLC7A5, resulting in bacterial escape from autophagy. These results expand our understanding of the mechanisms underlying the AA starvation response in Salmonella-infected cells.

18.
J Clin Gastroenterol ; 46 Suppl: S6-11, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22955360

ABSTRACT

AIM: To test the activities of culture-extracted or commercially available toll-like receptors (TLRs) ligands to establish their direct impact on target gastrointestinal motor cells. METHODS: Short-term and long-term effects of Shigella flexneri M90T and Escherichia coli K-2 strains-extracted lipopolysaccharides (LPS), commercially highly purified LPS (E. coli O111:B4 and EH100), and Pam2CSK4 and Pam3CSK4, which bind TLR2/6 and TLR1/2 heterodimers, respectively, have been assessed on pure primary cultures of colonic human smooth muscle cells (HSMC). RESULTS: Pathogenic Shigella-LPS and nonpathogenic E. coli K-2-LPS induced a time-dependent decrease of resting cell length and acetylcholine-induced contraction, with both alterations occurring rapidly and being more pronounced in response to the former. However, their effects differed, prolonging HSMC exposure with Shigella-LPS effects maintained throughout the 4 hours of observation compared with E. coli K-2-LPS, which disappeared after 60 minutes of incubation. Similar differences in magnitude and time dependency of myogenic effects were observed between pure TLR4 and TLR2/1 or TLR2/6 ligands. The specific activation of TLR4 with LPS from pathogen or nonpathogen E. coli, O111:B4 and EH100, respectively, induced smooth muscle alterations that progressively increased, prolonging incubation, whereas TLR2 ligands induced short-term alterations, of a lesser magnitude, which decreased over time. The real-time polymerase chain reaction analysis showed that HSMC express mRNA for TLR1, 2, 4, and 6, substantiating a direct effect of TLR ligands on human colonic smooth muscle. CONCLUSIONS: This study highlights that bacterial products can directly affect gastrointestinal motility and that TLRs subtypes may differ in their cellular activity.


Subject(s)
Escherichia coli/immunology , Gastrointestinal Motility/drug effects , Lipopolysaccharides/pharmacology , Myocytes, Smooth Muscle/drug effects , Shigella flexneri/immunology , Toll-Like Receptors/metabolism , Cells, Cultured , Colon/cytology , Colon/immunology , Colon/metabolism , Colon/physiopathology , Escherichia coli/metabolism , Gastrointestinal Motility/physiology , Humans , Immunity, Innate , Ligands , Metagenome , Muscle Contraction , Myocytes, Smooth Muscle/immunology , Myocytes, Smooth Muscle/metabolism , RNA, Messenger/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Shigella flexneri/metabolism , Toll-Like Receptor 2/drug effects , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/drug effects , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Toll-Like Receptors/drug effects , Toll-Like Receptors/genetics
19.
Autophagy ; 8(12): 1848-50, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22932645

ABSTRACT

Autophagy is a vital process through which cellular material and dysfunctional organelles are degraded and recycled, and it is inhibited by the metabolic checkpoint kinase MTOR. Autophagy also targets intracellular bacteria (a process termed xenophagy) for lysosomal degradation, thereby playing a key role in innate immunity. In the past few years, the identification of molecules, such as CALCOCO2/NDP52, SQSTM1/p62 and ubiquitin, implicated in the specific targeting of intracellular bacteria, received considerable attention. However, it remains unclear how xenophagy is initiated, since this process commonly occurs in metabolically replete cells. In a recent study, we demonstrated that infection with Shigella and Salmonella triggered an early state of intracellular amino acid (AA) starvation causing MTOR dissociation from endomembranes, downregulation of MTOR activity and activation of the EIF2AK4/GCN2-EIF2S1/eIF2α/ATF3 signaling axis. We also observed that AA starvation was caused by host membrane damage, which appeared to be transient in the case of Salmonella and sustained in Shigella-infected cells, thus highlighting the existence of key timing disparities in xenophagy triggering, depending on the bacterial pathogen. Together, our findings demonstrate that xenophagy is only one arm of a more general metabolic switch geared toward AA starvation in bacteria-infected cells.


Subject(s)
Autophagy , Salmonella/immunology , Shigella/immunology , Amino Acids/deficiency , Epithelial Cells/microbiology , Humans , Intracellular Space/microbiology , Models, Biological , Signal Transduction , Time Factors
20.
Cell Host Microbe ; 11(6): 563-75, 2012 Jun 14.
Article in English | MEDLINE | ID: mdl-22704617

ABSTRACT

Autophagy, which targets cellular constituents for degradation, is normally inhibited in metabolically replete cells by the metabolic checkpoint kinase mTOR. Although autophagic degradation of invasive bacteria has emerged as a critical host defense mechanism, the signals that induce autophagy upon bacterial infection remain unclear. We find that infection of epithelial cells with Shigella and Salmonella triggers acute intracellular amino acid (AA) starvation due to host membrane damage. Pathogen-induced AA starvation caused downregulation of mTOR activity, resulting in the induction of autophagy. In Salmonella-infected cells, membrane integrity and cytosolic AA levels rapidly normalized, favoring mTOR reactivation at the surface of the Salmonella-containing vacuole and bacterial escape from autophagy. In addition, bacteria-induced AA starvation activated the GCN2 kinase, eukaryotic initiation factor 2α, and the transcription factor ATF3-dependent integrated stress response and transcriptional reprogramming. Thus, AA starvation induced by bacterial pathogens is sensed by the host to trigger protective innate immune and stress responses.


Subject(s)
Amino Acids/metabolism , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Immunity, Innate , Salmonella/immunology , Shigella/immunology , Autophagy , Down-Regulation , Gene Expression Regulation , HeLa Cells , Humans , Salmonella/pathogenicity , Shigella/pathogenicity , TOR Serine-Threonine Kinases/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...