Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 11.117
Filter
Add more filters

Publication year range
1.
Cell ; 180(1): 50-63.e12, 2020 01 09.
Article in English | MEDLINE | ID: mdl-31923399

ABSTRACT

Mucosal barrier immunity is essential for the maintenance of the commensal microflora and combating invasive bacterial infection. Although immune and epithelial cells are thought to be the canonical orchestrators of this complex equilibrium, here, we show that the enteric nervous system (ENS) plays an essential and non-redundant role in governing the antimicrobial protein (AMP) response. Using confocal microscopy and single-molecule fluorescence in situ mRNA hybridization (smFISH) studies, we observed that intestinal neurons produce the pleiotropic cytokine IL-18. Strikingly, deletion of IL-18 from the enteric neurons alone, but not immune or epithelial cells, rendered mice susceptible to invasive Salmonella typhimurium (S.t.) infection. Mechanistically, unbiased RNA sequencing and single-cell sequencing revealed that enteric neuronal IL-18 is specifically required for homeostatic goblet cell AMP production. Together, we show that neuron-derived IL-18 signaling controls tissue-wide intestinal immunity and has profound consequences on the mucosal barrier and invasive bacterial killing.


Subject(s)
Immunity, Mucosal/immunology , Interleukin-18/immunology , Intestinal Mucosa/immunology , Animals , Cytokines/immunology , Enteric Nervous System/immunology , Enteric Nervous System/metabolism , Epithelial Cells/immunology , Female , Goblet Cells/immunology , Interleukin-18/biosynthesis , Intestinal Mucosa/metabolism , Intestine, Small/immunology , Male , Mice , Mice, Inbred C57BL , Neurons/immunology , Rats , Rats, Sprague-Dawley , Salmonella Infections/immunology , Salmonella typhimurium/immunology , Signal Transduction/immunology
2.
Cell ; 180(1): 33-49.e22, 2020 01 09.
Article in English | MEDLINE | ID: mdl-31813624

ABSTRACT

Gut-innervating nociceptor sensory neurons respond to noxious stimuli by initiating protective responses including pain and inflammation; however, their role in enteric infections is unclear. Here, we find that nociceptor neurons critically mediate host defense against the bacterial pathogen Salmonella enterica serovar Typhimurium (STm). Dorsal root ganglia nociceptors protect against STm colonization, invasion, and dissemination from the gut. Nociceptors regulate the density of microfold (M) cells in ileum Peyer's patch (PP) follicle-associated epithelia (FAE) to limit entry points for STm invasion. Downstream of M cells, nociceptors maintain levels of segmentous filamentous bacteria (SFB), a gut microbe residing on ileum villi and PP FAE that mediates resistance to STm infection. TRPV1+ nociceptors directly respond to STm by releasing calcitonin gene-related peptide (CGRP), a neuropeptide that modulates M cells and SFB levels to protect against Salmonella infection. These findings reveal a major role for nociceptor neurons in sensing and defending against enteric pathogens.


Subject(s)
Gastrointestinal Microbiome/physiology , Host Microbial Interactions/physiology , Nociceptors/physiology , Animals , Epithelium/metabolism , Female , Ganglia, Spinal/metabolism , Ganglia, Spinal/microbiology , Intestinal Mucosa/microbiology , Male , Mice , Mice, Inbred C57BL , Nociceptors/metabolism , Peyer's Patches/innervation , Peyer's Patches/metabolism , Salmonella Infections/metabolism , Salmonella typhimurium/metabolism , Salmonella typhimurium/pathogenicity , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/physiology
3.
Cell ; 183(3): 650-665.e15, 2020 10 29.
Article in English | MEDLINE | ID: mdl-33031742

ABSTRACT

Endocannabinoids are host-derived lipid hormones that fundamentally impact gastrointestinal (GI) biology. The use of cannabis and other exocannabinoids as anecdotal treatments for various GI disorders inspired the search for mechanisms by which these compounds mediate their effects, which led to the discovery of the mammalian endocannabinoid system. Dysregulated endocannabinoid signaling was linked to inflammation and the gut microbiota. However, the effects of endocannabinoids on host susceptibility to infection has not been explored. Here, we show that mice with elevated levels of the endocannabinoid 2-arachidonoyl glycerol (2-AG) are protected from enteric infection by Enterobacteriaceae pathogens. 2-AG directly modulates pathogen function by inhibiting virulence programs essential for successful infection. Furthermore, 2-AG antagonizes the bacterial receptor QseC, a histidine kinase encoded within the core Enterobacteriaceae genome that promotes the activation of pathogen-associated type three secretion systems. Taken together, our findings establish that endocannabinoids are directly sensed by bacteria and can modulate bacterial function.


Subject(s)
Endocannabinoids/metabolism , Enterobacteriaceae/pathogenicity , Animals , Arachidonic Acids/chemistry , Arachidonic Acids/metabolism , Bacterial Adhesion , Bacterial Proteins/metabolism , Bacterial Secretion Systems/metabolism , Citrobacter rodentium/pathogenicity , Colon/microbiology , Colon/pathology , Endocannabinoids/chemistry , Enterobacteriaceae Infections/microbiology , Female , Gastrointestinal Microbiome , Glycerides/chemistry , Glycerides/metabolism , HeLa Cells , Host-Pathogen Interactions , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Monoacylglycerol Lipases/metabolism , Salmonella/pathogenicity , Virulence
4.
Cell ; 173(4): 920-933.e13, 2018 05 03.
Article in English | MEDLINE | ID: mdl-29576451

ABSTRACT

Inflammasome activation is critical for host defenses against various microbial infections. Activation of the NLRC4 inflammasome requires detection of flagellin or type III secretion system (T3SS) components by NLR family apoptosis inhibitory proteins (NAIPs); yet how this pathway is regulated is unknown. Here, we found that interferon regulatory factor 8 (IRF8) is required for optimal activation of the NLRC4 inflammasome in bone-marrow-derived macrophages infected with Salmonella Typhimurium, Burkholderia thailandensis, or Pseudomonas aeruginosa but is dispensable for activation of the canonical and non-canonical NLRP3, AIM2, and Pyrin inflammasomes. IRF8 governs the transcription of Naips to allow detection of flagellin or T3SS proteins to mediate NLRC4 inflammasome activation. Furthermore, we found that IRF8 confers protection against bacterial infection in vivo, owing to its role in inflammasome-dependent cytokine production and pyroptosis. Altogether, our findings suggest that IRF8 is a critical regulator of NAIPs and NLRC4 inflammasome activation for defense against bacterial infection.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Calcium-Binding Proteins/metabolism , Inflammasomes/metabolism , Interferon Regulatory Factors/metabolism , Neuronal Apoptosis-Inhibitory Protein/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Calcium-Binding Proteins/genetics , Cells, Cultured , Cytokines/metabolism , Electrophoretic Mobility Shift Assay , Flagellin/metabolism , Interferon Regulatory Factors/antagonists & inhibitors , Interferon Regulatory Factors/genetics , Macrophages/cytology , Macrophages/metabolism , Macrophages/microbiology , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neuronal Apoptosis-Inhibitory Protein/genetics , Promoter Regions, Genetic , Protein Binding , Pseudomonas aeruginosa/pathogenicity , Pyroptosis , RNA Interference , RNA, Small Interfering/metabolism , Salmonella typhimurium/pathogenicity , Transcription, Genetic
5.
Cell ; 168(6): 1065-1074.e10, 2017 03 09.
Article in English | MEDLINE | ID: mdl-28283062

ABSTRACT

Type III protein secretion systems have specifically evolved to deliver bacterially encoded proteins into target eukaryotic cells. The core elements of this multi-protein machine are the envelope-associated needle complex, the inner membrane export apparatus, and a large cytoplasmic sorting platform. Here, we report a high-resolution in situ structure of the Salmonella Typhimurium type III secretion machine obtained by high-throughput cryo-electron tomography and sub-tomogram averaging. Through molecular modeling and comparative analysis of machines assembled with protein-tagged components or from different deletion mutants, we determined the molecular architecture of the secretion machine in situ and localized its structural components. We also show that docking of the sorting platform results in significant conformational changes in the needle complex to provide the symmetry adaptation required for the assembly of the entire secretion machine. These studies provide major insight into the structure and assembly of a broadly distributed protein secretion machine.


Subject(s)
Bacterial Secretion Systems/ultrastructure , Salmonella typhimurium/ultrastructure , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Secretion Systems/genetics , Cryoelectron Microscopy , Protein Transport , Virulence
6.
Cell ; 168(3): 503-516.e12, 2017 01 26.
Article in English | MEDLINE | ID: mdl-28129542

ABSTRACT

Sickness-induced anorexia is a conserved behavior induced during infections. Here, we report that an intestinal pathogen, Salmonella Typhimurium, inhibits anorexia by manipulating the gut-brain axis. Inhibition of inflammasome activation by the S. Typhimurium effector, SlrP, prevented anorexia caused by IL-1ß-mediated signaling to the hypothalamus via the vagus nerve. Rather than compromising host defenses, pathogen-mediated inhibition of anorexia increased host survival. SlrP-mediated inhibition of anorexia prevented invasion and systemic infection by wild-type S. Typhimurium, reducing virulence while increasing transmission to new hosts, suggesting that there are trade-offs between transmission and virulence. These results clarify the complex and contextual role of anorexia in host-pathogen interactions and suggest that microbes have evolved mechanisms to modulate sickness-induced behaviors to promote health of their host and their transmission at the expense of virulence.


Subject(s)
Anorexia/microbiology , Salmonella Infections/microbiology , Salmonella Infections/transmission , Salmonella typhimurium/pathogenicity , Animals , Bacterial Proteins/metabolism , Host-Pathogen Interactions , Humans , Inflammasomes/immunology , Interleukin-1beta/immunology , Mice , Mice, Inbred C57BL , Neural Pathways , Salmonella Infections/immunology , Salmonella typhimurium/physiology , Specific Pathogen-Free Organisms , Virulence
7.
Immunity ; 55(3): 442-458.e8, 2022 03 08.
Article in English | MEDLINE | ID: mdl-35182483

ABSTRACT

Consecutive exposures to different pathogens are highly prevalent and often alter the host immune response. However, it remains unknown how a secondary bacterial infection affects an ongoing adaptive immune response elicited against primary invading pathogens. We demonstrated that recruitment of Sca-1+ monocytes into lymphoid organs during Salmonella Typhimurium (STm) infection disrupted pre-existing germinal center (GC) reactions. GC responses induced by influenza, plasmodium, or commensals deteriorated following STm infection. GC disruption was independent of the direct bacterial interactions with B cells and instead was induced through recruitment of CCR2-dependent Sca-1+ monocytes into the lymphoid organs. GC collapse was associated with impaired cellular respiration and was dependent on TNFα and IFNγ, the latter of which was essential for Sca-1+ monocyte differentiation. Monocyte recruitment and GC disruption also occurred during LPS-supplemented vaccination and Listeria monocytogenes infection. Thus, systemic activation of the innate immune response upon severe bacterial infection is induced at the expense of antibody-mediated immunity.


Subject(s)
Bacterial Infections , Listeriosis , B-Lymphocytes , Germinal Center , Humans , Monocytes
8.
Mol Cell ; 83(24): 4538-4554.e4, 2023 Dec 21.
Article in English | MEDLINE | ID: mdl-38091999

ABSTRACT

Homologous to E6AP C terminus (HECT) E3 ubiquitin (Ub) ligases direct substrates toward distinct cellular fates dictated by the specific form of monomeric or polymeric Ub (polyUb) signal attached. How polyUb specificity is achieved has been a long-standing mystery, despite extensive study in various hosts, ranging from yeast to human. The bacterial pathogens enterohemorrhagic Escherichia coli and Salmonella Typhimurium encode outlying examples of "HECT-like" (bHECT) E3 ligases, but commonalities to eukaryotic HECT (eHECT) mechanism and specificity had not been explored. We expanded the bHECT family with examples in human and plant pathogens. Three bHECT structures in primed, Ub-loaded states resolved key details of the entire Ub ligation process. One structure provided a rare glimpse into the act of ligating polyUb, yielding a means to rewire polyUb specificity of both bHECT and eHECT ligases. Studying this evolutionarily distinct bHECT family has revealed insight into the function of key bacterial virulence factors as well as fundamental principles underlying HECT-type Ub ligation.


Subject(s)
Polyubiquitin , Ubiquitin-Protein Ligases , Humans , Polyubiquitin/genetics , Polyubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
9.
Immunity ; 54(4): 687-701.e4, 2021 04 13.
Article in English | MEDLINE | ID: mdl-33773107

ABSTRACT

Interferon-γ (IFN-γ)-producing CD4+ T helper-1 (Th1) cells are critical for protection from microbes that infect the phagosomes of myeloid cells. Current understanding of Th1 cell differentiation is based largely on reductionist cell culture experiments. We assessed Th1 cell generation in vivo by studying antigen-specific CD4+ T cells during infection with the phagosomal pathogen Salmonella enterica (Se), or influenza A virus (IAV), for which CD4+ T cells are less important. Both microbes induced T follicular helper (Tfh) and interleukin-12 (IL-12)-independent Th1 cells. During Se infection, however, the Th1 cells subsequently outgrew the Tfh cells via an IL-12-dependent process and formed subsets with increased IFN-γ production, ZEB2-transcription factor-dependent cytotoxicity, and capacity to control Se infection. Our results indicate that many infections induce a module that generates Tfh and poorly differentiated Th1 cells, which is followed in phagosomal infections by an IL-12-dependent Th1 cell amplification module that is critical for pathogen control.


Subject(s)
Cell Differentiation/immunology , Th1 Cells/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Line , Drosophila/immunology , Female , Interferon-gamma/immunology , Interleukin-12/immunology , Lymphocyte Activation/immunology , Male , Mice, Inbred C57BL , T-Lymphocytes, Helper-Inducer/immunology
10.
Immunity ; 54(12): 2712-2723.e6, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34788598

ABSTRACT

Interactions between intracellular bacteria and mononuclear phagocytes give rise to diverse cellular phenotypes that may determine the outcome of infection. Recent advances in single-cell RNA sequencing (scRNA-seq) have identified multiple subsets within the mononuclear population, but implications to their function during infection are limited. Here, we surveyed the mononuclear niche of intracellular Salmonella Typhimurium (S.Tm) during early systemic infection in mice. We described eclipse-like growth kinetics in the spleen, with a first phase of bacterial control mediated by tissue-resident red-pulp macrophages. A second phase involved extensive bacterial replication within a macrophage population characterized by CD9 expression. We demonstrated that CD9+ macrophages induced pathways for detoxificating oxidized lipids, that may be utilized by intracellular S.Tm. We established that CD9+ macrophages originated from non-classical monocytes (NCM), and NCM-depleted mice were more resistant to S.Tm infection. Our study defines macrophage subset-specific host-pathogen interactions that determine early infection dynamics and infection outcome of the entire organism.


Subject(s)
Macrophages/immunology , Salmonella Infections/immunology , Salmonella typhimurium/physiology , Spleen/immunology , Animals , Host-Pathogen Interactions , Humans , Intracellular Space , Lipid Metabolism , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oxidation-Reduction , Single-Cell Analysis , Spleen/microbiology , Tetraspanin 29/metabolism
11.
Mol Cell ; 82(3): 629-644.e4, 2022 02 03.
Article in English | MEDLINE | ID: mdl-35063132

ABSTRACT

The envelope of Gram-negative bacteria is a vital barrier that must balance protection and nutrient uptake. Small RNAs are crucial regulators of the envelope composition and function. Here, using RIL-seq to capture the Hfq-mediated RNA-RNA interactome in Salmonella enterica, we discover envelope-related riboregulators, including OppX. We show that OppX acts as an RNA sponge of MicF sRNA, a prototypical porin repressor. OppX originates from the 5' UTR of oppABCDF, encoding the major inner-membrane oligopeptide transporter, and sequesters MicF's seed region to derepress the synthesis of the porin OmpF. Intriguingly, OppX operates as a true sponge, storing MicF in an inactive complex without affecting its levels or stability. Conservation of the opp-OppX-MicF-ompF axis in related bacteria suggests that it serves an important mechanism, adjusting envelope porosity to specific transport capacity. These data also highlight the resource value of this Salmonella RNA interactome, which will aid in unraveling RNA-centric regulation in enteric pathogens.


Subject(s)
5' Untranslated Regions , Cell Membrane/genetics , Escherichia coli Proteins/genetics , Host Factor 1 Protein/genetics , RNA, Bacterial/genetics , Salmonella enterica/genetics , Biological Transport , Cell Membrane/metabolism , Escherichia coli Proteins/metabolism , Gene Expression Regulation, Bacterial , Host Factor 1 Protein/metabolism , Host-Pathogen Interactions , Permeability , Porins/genetics , Porins/metabolism , RNA, Bacterial/metabolism , RNA-Seq , Salmonella enterica/metabolism , Salmonella enterica/pathogenicity
12.
Immunity ; 53(3): 533-547.e7, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32735843

ABSTRACT

Programmed cell death contributes to host defense against pathogens. To investigate the relative importance of pyroptosis, necroptosis, and apoptosis during Salmonella infection, we infected mice and macrophages deficient for diverse combinations of caspases-1, -11, -12, and -8 and receptor interacting serine/threonine kinase 3 (RIPK3). Loss of pyroptosis, caspase-8-driven apoptosis, or necroptosis had minor impact on Salmonella control. However, combined deficiency of these cell death pathways caused loss of bacterial control in mice and their macrophages, demonstrating that host defense can employ varying components of several cell death pathways to limit intracellular infections. This flexible use of distinct cell death pathways involved extensive cross-talk between initiators and effectors of pyroptosis and apoptosis, where initiator caspases-1 and -8 also functioned as executioners when all known effectors of cell death were absent. These findings uncover a highly coordinated and flexible cell death system with in-built fail-safe processes that protect the host from intracellular infections.


Subject(s)
Apoptosis/immunology , Macrophages/immunology , Necroptosis/immunology , Pyroptosis/immunology , Salmonella Infections/immunology , Salmonella/immunology , Animals , Caspase 1/deficiency , Caspase 1/genetics , Caspase 12/deficiency , Caspase 12/genetics , Caspase 8/genetics , Caspases, Initiator/deficiency , Caspases, Initiator/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Receptor-Interacting Protein Serine-Threonine Kinases/genetics
13.
EMBO J ; 43(8): 1499-1518, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38528181

ABSTRACT

The intestinal pathogen Salmonella enterica rapidly enters the bloodstream after the invasion of intestinal epithelial cells, but how Salmonella breaks through the gut-vascular barrier is largely unknown. Here, we report that Salmonella enters the bloodstream through intestinal CX3CR1+ macrophages during early infection. Mechanistically, Salmonella induces the migration/invasion properties of macrophages in a manner dependent on host cell actin and on the pathogen effector SteC. SteC recruits host myosin light chain protein Myl12a and phosphorylates its Ser19 and Thr20 residues. Myl12a phosphorylation results in actin rearrangement, and enhanced migration and invasion of macrophages. SteC is able to utilize a wide range of NTPs other than ATP to phosphorylate Myl12a. We further solved the crystal structure of SteC, which suggests an atypical dimerization-mediated catalytic mechanism. Finally, in vivo data show that SteC-mediated cytoskeleton manipulation is crucial for Salmonella breaching the gut vascular barrier and spreading to target organs.


Subject(s)
Myosin Light Chains , Salmonella enterica , Myosin Light Chains/genetics , Myosin Light Chains/metabolism , Actins/metabolism , Epithelial Cells/metabolism , Macrophages/metabolism
14.
Annu Rev Microbiol ; 77: 645-668, 2023 09 15.
Article in English | MEDLINE | ID: mdl-37713459

ABSTRACT

Secretory antibodies are the only component of our adaptive immune system capable of attacking mucosal pathogens topologically outside of our bodies. All secretory antibody classes are (a) relatively resistant to harsh proteolytic environments and (b) polymeric. Recent elucidation of the structure of secretory IgA (SIgA) has begun to shed light on SIgA functions at the nanoscale. We can now begin to unravel the structure-function relationships of these molecules, for example, by understanding how the bent conformation of SIgA enables robust cross-linking between adjacent growing bacteria. Many mysteries remain, such as the structural basis of protease resistance and the role of noncanonical bacteria-IgA interactions. In this review, we explore the structure-function relationships of IgA from the nano- to the metascale, with a strong focus on how the seemingly banal "license to clump" can have potent effects on bacterial physiology and colonization.


Subject(s)
Immunoglobulin A, Secretory , Biological Transport , Structure-Activity Relationship
15.
Annu Rev Cell Dev Biol ; 30: 79-109, 2014.
Article in English | MEDLINE | ID: mdl-25103867

ABSTRACT

Pathogens use a vast number of strategies to alter host membrane dynamics. Targeting the host membrane machinery is important for the survival and pathogenesis of several extracellular, vacuolar, and cytosolic bacteria. Membrane manipulation promotes bacterial replication while suppressing host responses, allowing the bacterium to thrive in a hostile environment. This review provides a comprehensive summary of various strategies used by both extracellular and intracellular bacteria to hijack host membrane trafficking machinery. We start with mechanisms used by bacteria to alter the plasma membrane, delve into the hijacking of various vesicle trafficking pathways, and conclude by summarizing bacterial adaptation to host immune responses. Understanding bacterial manipulation of host membrane trafficking provides insights into bacterial pathogenesis and uncovers the molecular mechanisms behind various processes within a eukaryotic cell.


Subject(s)
Bacterial Physiological Phenomena , Cell Membrane/metabolism , Cells/microbiology , Host-Pathogen Interactions/physiology , Animals , Autophagy/physiology , Bacterial Proteins/physiology , Bacterial Toxins/pharmacology , Biological Transport , Cell Membrane Permeability , Cells/ultrastructure , Cytosol/microbiology , Endocytosis/physiology , Humans , Lysosomes/physiology , Membrane Lipids/metabolism , Membrane Proteins/metabolism , Phagosomes/physiology , Protein Transport , Vacuoles/microbiology , Vacuoles/physiology
16.
EMBO J ; 42(13): e112333, 2023 07 03.
Article in English | MEDLINE | ID: mdl-37183585

ABSTRACT

Enteric bacteria use up to 15% of their cellular energy for ammonium assimilation via glutamine synthetase (GS)/glutamate synthase (GOGAT) and glutamate dehydrogenase (GDH) in response to varying ammonium availability. However, the sensory mechanisms for effective and appropriate coordination between carbon metabolism and ammonium assimilation have not been fully elucidated. Here, we report that in Salmonella enterica, carbon metabolism coordinates the activities of GS/GDH via functionally reversible protein lysine acetylation. Glucose promotes Pat acetyltransferase-mediated acetylation and activation of adenylylated GS. Simultaneously, glucose induces GDH acetylation to inactivate the enzyme by impeding its catalytic centre, which is reversed upon GDH deacetylation by deacetylase CobB. Molecular dynamics (MD) simulations indicate that adenylylation is required for acetylation-dependent activation of GS. We show that acetylation and deacetylation occur within minutes of "glucose shock" to promptly adapt to ammonium/carbon variation and finely balance glutamine/glutamate synthesis. Finally, in a mouse infection model, reduced S. enterica growth caused by the expression of adenylylation-mimetic GS is rescued by acetylation-mimicking mutations. Thus, glucose-driven acetylation integrates signals from ammonium assimilation and carbon metabolism to fine-tune bacterial growth control.


Subject(s)
Ammonium Compounds , Salmonella enterica , Animals , Mice , Ammonium Compounds/metabolism , Acetylation , Carbon/metabolism , Glucose , Glutamate Dehydrogenase/metabolism , Nitrogen/metabolism
17.
Immunity ; 49(1): 151-163.e5, 2018 07 17.
Article in English | MEDLINE | ID: mdl-29980437

ABSTRACT

The intestinal barrier is vulnerable to damage by microbiota-induced inflammation that is normally restrained through mechanisms promoting homeostasis. Such disruptions contribute to autoimmune and inflammatory diseases including inflammatory bowel disease. We identified a regulatory loop whereby, in the presence of the normal microbiota, intestinal antigen-presenting cells (APCs) expressing the chemokine receptor CX3CR1 reduced expansion of intestinal microbe-specific T helper 1 (Th1) cells and promoted generation of regulatory T cells responsive to food antigens and the microbiota itself. We identified that disruption of the microbiota resulted in CX3CR1+ APC-dependent inflammatory Th1 cell responses with increased pathology after pathogen infection. Colonization with microbes that can adhere to the epithelium was able to compensate for intestinal microbiota loss, indicating that although microbial interactions with the epithelium can be pathogenic, they can also activate homeostatic regulatory mechanisms. Our results identify a cellular mechanism by which the microbiota limits intestinal inflammation and promotes tissue homeostasis.


Subject(s)
CX3C Chemokine Receptor 1/metabolism , Gastrointestinal Microbiome/immunology , Intestinal Mucosa/immunology , Mononuclear Phagocyte System/immunology , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Animals , Antigen Presentation , Bacterial Adhesion/immunology , Disease Models, Animal , Female , Homeostasis , Immune Tolerance , Immunity, Mucosal , Inflammation/immunology , Inflammatory Bowel Diseases/immunology , Interleukin-10/immunology , Interleukin-10/metabolism , Intestinal Mucosa/microbiology , Male , Mice , RAW 264.7 Cells
18.
Mol Cell ; 74(2): 320-329.e6, 2019 04 18.
Article in English | MEDLINE | ID: mdl-30853402

ABSTRACT

Xenophagy, a selective autophagy pathway that protects the cytosol against bacterial invasion, relies on cargo receptors that juxtapose bacteria and phagophore membranes. Whether phagophores are recruited from a constitutive pool or are generated de novo at prospective cargo remains unknown. Phagophore formation in situ would require recruitment of the upstream autophagy machinery to prospective cargo. Here, we show that, essential for anti-bacterial autophagy, the cargo receptor NDP52 forms a trimeric complex with FIP200 and SINTBAD/NAP1, which are subunits of the autophagy-initiating ULK and the TBK1 kinase complex, respectively. FIP200 and SINTBAD/NAP1 are each recruited independently to bacteria via NDP52, as revealed by selective point mutations in their respective binding sites, but only in their combined presence does xenophagy proceed. Such recruitment of the upstream autophagy machinery by NDP52 reveals how detection of cargo-associated "eat me" signals, induction of autophagy, and juxtaposition of cargo and phagophores are integrated in higher eukaryotes.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Autophagy/genetics , Nuclear Proteins/genetics , Protein-Tyrosine Kinases/genetics , Adaptor Proteins, Signal Transducing/chemistry , Autophagy-Related Protein-1 Homolog/genetics , Autophagy-Related Proteins , Binding Sites/genetics , Cytoplasm/microbiology , Cytosol/microbiology , Humans , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Nuclear Proteins/chemistry , Point Mutation/genetics , Protein Binding/genetics , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/chemistry , Salmonella typhimurium/genetics , Salmonella typhimurium/pathogenicity
19.
Semin Immunol ; 70: 101812, 2023 11.
Article in English | MEDLINE | ID: mdl-37562110

ABSTRACT

Besides its crucial function in nutrient absorbance and as barrier against the microbiota, the gut epithelium is essential for sensing pathogenic insults and mounting of an appropriate early immune response. In mice, the activation of the canonical NAIP/NLRC4 inflammasome is critical for the defense against enterobacterial infections. Activation of the NAIP/NLRC4 inflammasome triggers the extrusion of infected intestinal epithelial cells (IEC) into the gut lumen, concomitant with inflammasome-mediated lytic cell death. The membrane permeabilization, a hallmark of pyroptosis, is caused by the pore-forming proteins called gasdermins (GSDMs). Recent work has revealed that NAIP/NLRC4-dependent extrusion of infected IECs can, however, also be executed in the absence of GSDMD. In fact, several reports highlighted that various cell death pathways (e.g., pyroptosis or apoptosis) and unique mechanisms specific to particular infection models and stages of gut infection are in action during epithelial inflammasome defense against intestinal pathogens. Here, we summarize the current knowledge regarding the underlying mechanisms and speculate on the putative functions of the epithelial inflammasome activation and cell death, with a particular emphasis on mouse infection models for two prominent enterobacterial pathogens, Salmonella Typhimurium and Shigella flexneri.


Subject(s)
Inflammasomes , Shigella , Mice , Animals , Humans , Gasdermins , Salmonella typhimurium/metabolism , Shigella/metabolism , Inflammation
20.
EMBO Rep ; 25(2): 704-724, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38263327

ABSTRACT

TFEB is a master regulator of autophagy, lysosome biogenesis, mitochondrial metabolism, and immunity that works primarily through transcription controlled by cytosol-to-nuclear translocation. Emerging data indicate additional regulatory interactions at the surface of organelles such as lysosomes. Here we show that TFEB has a non-transcriptional role in mitochondria, regulating the electron transport chain complex I to down-modulate inflammation. Proteomics analysis reveals extensive TFEB co-immunoprecipitation with several mitochondrial proteins, whose interactions are disrupted upon infection with S. Typhimurium. High resolution confocal microscopy and biochemistry confirms TFEB localization in the mitochondrial matrix. TFEB translocation depends on a conserved N-terminal TOMM20-binding motif and is enhanced by mTOR inhibition. Within the mitochondria, TFEB and protease LONP1 antagonistically co-regulate complex I, reactive oxygen species and the inflammatory response. Consequently, during infection, lack of TFEB specifically in the mitochondria exacerbates the expression of pro-inflammatory cytokines, contributing to innate immune pathogenesis.


Subject(s)
Autophagy , Inflammation , Humans , Inflammation/metabolism , Cytosol/metabolism , Active Transport, Cell Nucleus , Lysosomes/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Mitochondrial Proteins/metabolism , ATP-Dependent Proteases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL