Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Bioinformatics ; 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38991828

ABSTRACT

MOTIVATION: Sanger sequencing of taxonomic marker genes (e.g., 16S/18S/ITS/rpoB/cpn60) represents the leading method for identifying a wide range of microorganisms including bacteria, archaea, and fungi. However, the manual processing of sequence data and limitations associated with conventional BLAST searches impede the efficient generation of strain libraries essential for cataloging microbial diversity and discovering novel species. RESULTS: isolateR addresses these challenges by implementing a standardized and scalable three-step pipeline that includes: 1) automated batch processing of Sanger sequence files, 2) taxonomic classification via global alignment to type strain databases in accordance with the latest international nomenclature standards, and 3) straightforward creation of strain libraries and handling of clonal isolates, with the ability to set customizable sequence dereplication thresholds and combine data from multiple sequencing runs into a single library. The tool's user-friendly design also features interactive HTML outputs that simplify data exploration and analysis. Additionally, in silico benchmarking done on two comprehensive human gut genome catalogues (IMGG and Hadza hunter-gather populations) showcase the proficiency of isolateR in uncovering and cataloging the nuanced spectrum of microbial diversity, advocating for a more targeted and granular exploration within individual hosts to achieve the highest strain-level resolution possible when generating culture collections. AVAILABILITY: isolateR is available at: https://github.com/bdaisley/isolateR. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.

2.
Trends Endocrinol Metab ; 35(2): 91-93, 2024 02.
Article in English | MEDLINE | ID: mdl-37945457

ABSTRACT

In a recent article, Button and colleagues demonstrate that human milk oligosaccharides create a nutrient niche that supports reversible colonization by Bifidobacterium infantis. Using this tunable system, they assessed the impact of B. infantis on microbiome recovery after antibiotic treatment. Overall, this work highlights synbiotics as a useful approach for developing live biotherapeutic products (LBPs).


Subject(s)
Microbiota , Synbiotics , Humans , Milk, Human , Bifidobacterium , Oligosaccharides
3.
Cell Host Microbe ; 31(12): 1950-1951, 2023 Dec 13.
Article in English | MEDLINE | ID: mdl-38096786

ABSTRACT

Exclusive enteral nutrition, a diet lacking fiber, is used to treat pediatric Crohn's disease. In this issue of Cell Host & Microbe, Kuffa et al. find that a fiber-deficient diet thins the mucus layer and alters microbial cross-feeding, causing pro-inflammatory Mucispirillum to move away from the epithelium, which ameliorates colitis.


Subject(s)
Crohn Disease , Microbiota , Child , Humans , Crohn Disease/therapy , Enteral Nutrition , Diet , Dietary Fiber
4.
Environ Microbiol ; 24(7): 3111-3123, 2022 07.
Article in English | MEDLINE | ID: mdl-35466558

ABSTRACT

Species of the genus Blautia are typical inhabitants of the human gut and considered as beneficial gut microbes. However, their role in the gut microbiome and their metabolic features are poorly understood. Blautia schinkii was described as an acetogenic bacterium, characterized by a functional Wood-Ljungdahl pathway (WLP) of acetogenesis from H2  + CO2 . Here we report that two relatives, Blautia luti and Blautia wexlerae do not grow on H2  + CO2 . Inspection of the genome sequence revealed all genes of the WLP except genes encoding a formate dehydrogenase and an electron-bifurcating hydrogenase. Enzyme assays confirmed this prediction. Accordingly, resting cells neither converted H2  + CO2 nor H2  + HCOOH + CO2 to acetate. Carbon monoxide is an intermediate of the WLP and substrate for many acetogens. Blautia luti and B. wexlerae had an active CO dehydrogenase and resting cells performed acetogenesis from HCOOH + CO2  + CO, demonstrating a functional WLP. Bioinformatic analyses revealed that many Blautia strains as well as other gut acetogens lack formate dehydrogenases and hydrogenases. Thus, the use of formate instead of H2  + CO2 as an interspecies hydrogen and electron carrier seems to be more common in the gut microbiome.


Subject(s)
Formate Dehydrogenases , Hydrogenase , Bacterial Proteins/metabolism , Carbon Dioxide/metabolism , Clostridiales , Formate Dehydrogenases/genetics , Humans , Hydrogenase/genetics , Wood/metabolism
5.
Nat Rev Microbiol ; 20(6): 365-380, 2022 06.
Article in English | MEDLINE | ID: mdl-34992261

ABSTRACT

Symbiotic microorganisms inhabiting the gastrointestinal tract promote health by decreasing susceptibility to infection and enhancing resistance to a range of diseases. In this Review, we discuss our increasing understanding of the impact of the microbiome on the mammalian host and recent efforts to culture and characterize intestinal symbiotic microorganisms that produce or modify metabolites that impact disease pathology. Manipulation of the intestinal microbiome has great potential to reduce the incidence and/or severity of a wide range of human conditions and diseases, and the biomedical research community now faces the challenge of translating our understanding of the microbiome into beneficial medical therapies. Our increasing understanding of symbiotic microbial species and the application of ecological principles and machine learning are providing exciting opportunities for microbiome-based therapeutics to progress from faecal microbiota transplantation to the administration of precisely defined and clinically validated symbiotic microbial consortia that optimize disease resistance.


Subject(s)
Gastrointestinal Microbiome , Microbiota , Fecal Microbiota Transplantation , Gastrointestinal Tract , Health Promotion , Humans
6.
PLoS Pathog ; 17(2): e1009309, 2021 02.
Article in English | MEDLINE | ID: mdl-33556154

ABSTRACT

Gram-negative pathogens, such as Klebsiella pneumoniae, remodel their outer membrane (OM) in response to stress to maintain its integrity as an effective barrier and thus to promote their survival in the host. The emergence of carbapenem-resistant K. pneumoniae (CR-Kp) strains that are resistant to virtually all antibiotics is an increasing clinical problem and OM impermeability has limited development of antimicrobial agents because higher molecular weight antibiotics cannot access sites of activity. Here, we demonstrate that TAM (translocation and assembly module) deletion increases CR-Kp OM permeability under stress conditions and enhances sensitivity to high-molecular weight antimicrobials. SILAC-based proteomic analyses revealed mis-localization of membrane proteins in the TAM deficient strain. Stress-induced sensitization enhances clearance of TAM-deficient CR-Kp from the gut lumen following fecal microbiota transplantation and from infection sites following pulmonary or systemic infection. Our study suggests that TAM, as a regulator of OM permeability, represents a potential target for development of agents that enhance the effectiveness of existing antibiotics.


Subject(s)
Adaptation, Physiological , Anti-Bacterial Agents/pharmacology , Bacterial Outer Membrane Proteins/metabolism , Carbapenem-Resistant Enterobacteriaceae/drug effects , Klebsiella Infections/drug therapy , Klebsiella pneumoniae/drug effects , Proteome/metabolism , Animals , Bacterial Outer Membrane Proteins/genetics , Carbapenems/pharmacology , Cell Membrane Permeability , Female , Klebsiella Infections/genetics , Klebsiella Infections/metabolism , Klebsiella Infections/microbiology , Mice , Mice, Inbred C57BL , Stress, Physiological
7.
Cell Host Microbe ; 29(3): 378-393.e5, 2021 03 10.
Article in English | MEDLINE | ID: mdl-33539766

ABSTRACT

The gut microbiota produces metabolites that regulate host immunity, thereby impacting disease resistance and susceptibility. The extent to which commensal bacteria reciprocally respond to immune activation, however, remains largely unexplored. Herein, we colonized mice with four anaerobic symbionts and show that acute immune responses result in dramatic transcriptional reprogramming of these commensals with minimal changes in their relative abundance. Transcriptomic changes include induction of stress-response mediators and downregulation of carbohydrate-degrading factors such as polysaccharide utilization loci (PULs). Flagellin and anti-CD3 antibody, two distinct immune stimuli, induced similar transcriptional profiles, suggesting that commensal bacteria detect common effectors or activate shared pathways when facing different host responses. Immune activation altered the intestinal metabolome within 6 hours, decreasing luminal short-chain fatty acid and increasing aromatic metabolite concentrations. Thus, intestinal bacteria, prior to detectable shifts in community composition, respond to acute host immune activation by rapidly changing gene transcription and immunomodulatory metabolite production.


Subject(s)
Gastrointestinal Microbiome/immunology , Gastrointestinal Microbiome/physiology , Intestines/immunology , Intestines/microbiology , Animals , Bacteria/genetics , Bacteria/metabolism , Cross-Sectional Studies , Down-Regulation , Fatty Acids, Volatile , Female , Flagellin , Gastrointestinal Microbiome/genetics , Inflammation/immunology , Metabolome , Mice , Mice, Inbred C57BL , RNA, Ribosomal, 16S , Symbiosis , Transcriptome
8.
Cell Host Microbe ; 28(1): 134-146.e4, 2020 07 08.
Article in English | MEDLINE | ID: mdl-32492369

ABSTRACT

Bacteria belonging to the Lachnospiraceae family are abundant, obligate anaerobic members of the microbiota in healthy humans. Lachnospiraceae impact their hosts by producing short-chain fatty acids, converting primary to secondary bile acids, and facilitating colonization resistance against intestinal pathogens. To increase our understanding of genomic and functional diversity between members of this family, we cultured 273 Lachnospiraceae isolates representing 11 genera and 27 species from human donors and performed whole-genome sequencing assembly and annotation. This analysis revealed substantial inter- and intra-species diversity in pathways that likely influence an isolate's ability to impact host health. These differences are likely to impact colonization resistance through lantibiotic expression or intestinal acidification, influence host mucosal immune cells and enterocytes via butyrate production, or contribute to synergism within a consortium by heterogenous polysaccharide metabolism. Identification of these specific functions could facilitate development of probiotic bacterial consortia that drive and/or restore in vivo microbiome functions.


Subject(s)
Clostridiales/classification , Clostridiales/genetics , Gastrointestinal Microbiome/genetics , Genetic Variation , Metabolic Networks and Pathways/genetics , Feces/microbiology , Genome, Bacterial , Humans , Metagenomics , Phylogeny , RNA, Ribosomal, 16S/genetics , Whole Genome Sequencing
9.
Infect Immun ; 88(4)2020 03 23.
Article in English | MEDLINE | ID: mdl-31907198

ABSTRACT

Antibiotic treatment of patients undergoing complex medical treatments can deplete commensal bacterial strains from the intestinal microbiota, thereby reducing colonization resistance against a wide range of antibiotic-resistant pathogens. Loss of colonization resistance can lead to marked expansion of vancomycin-resistant Enterococcus faecium (VRE), Klebsiella pneumoniae, and Escherichia coli in the intestinal lumen, predisposing patients to bloodstream invasion and sepsis. The impact of intestinal domination by these antibiotic-resistant pathogens on mucosal immune defenses and epithelial and mucin-mediated barrier integrity is unclear. We used a mouse model to study the impact of intestinal domination by antibiotic-resistant bacterial species and strains on the colonic mucosa. Intestinal colonization with K. pneumoniae, Proteus mirabilis, or Enterobacter cloacae promoted greater recruitment of neutrophils to the colonic mucosa. To test the hypothesis that the residual microbiota influences the severity of colitis caused by infection with Clostridioides difficile, we coinfected mice that were colonized with ampicillin-resistant bacteria with a virulent strain of C. difficile and monitored colonization and pathogenesis. Despite the compositional differences in the gut microbiota, the severity of C. difficile infection (CDI) and mortality did not differ significantly between mice colonized with different ampicillin-resistant bacterial species. Our results suggest that the virulence mechanisms enabling CDI and epithelial destruction outweigh the relatively minor impact of less-virulent antibiotic-resistant intestinal bacteria on the outcome of CDI.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Clostridium Infections/physiopathology , Drug Resistance, Bacterial , Enterobacter cloacae/growth & development , Enterobacteriaceae Infections/complications , Klebsiella pneumoniae/growth & development , Proteus mirabilis/growth & development , Animals , Clostridium Infections/microbiology , Colitis/microbiology , Colitis/physiopathology , Disease Models, Animal , Enterobacter cloacae/drug effects , Enterobacteriaceae Infections/drug therapy , Klebsiella pneumoniae/drug effects , Mice , Microbial Interactions , Proteus mirabilis/drug effects , Survival Analysis
10.
Mol Cell Biol ; 39(10)2019 05 15.
Article in English | MEDLINE | ID: mdl-30910794

ABSTRACT

Protein degradation pathways are critical for maintaining proper protein dynamics within the cell, and considerable efforts have been made toward the development of therapeutics targeting these catabolic processes. We report here that isoginkgetin, a naturally derived biflavonoid, sensitized cells undergoing nutrient starvation to apoptosis, induced lysosomal stress, and activated the lysosome biogenesis gene TFEB Isoginkgetin treatment led to the accumulation of aggregates of polyubiquitinated proteins that colocalized strongly with the adaptor protein p62, the 20S proteasome, and the endoplasmic reticulum-associated degradation (ERAD) protein UFD1L. Isoginkgetin directly inhibited the chymotrypsin-like, trypsin-like, and caspase-like activities of the 20S proteasome and impaired NF-κB signaling, suggesting that the molecule may display its biological activity in part through proteasome inhibition. Importantly, isoginkgetin was effective at killing multiple myeloma (MM) cell lines in vitro and displayed a higher rate of cell death induction than the clinically approved proteasome inhibitor bortezomib. We propose that isoginkgetin disturbs protein homeostasis, leading to an excess of protein cargo that places a burden on the lysosomes/autophagic machinery, eventually leading to cancer cell death.


Subject(s)
Biflavonoids/pharmacology , Lysosomes/metabolism , Multiple Myeloma/metabolism , Proteasome Inhibitors/pharmacology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Line, Tumor , Cell Survival/drug effects , HCT116 Cells , HeLa Cells , Homeostasis/drug effects , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lysosomes/drug effects , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , NF-kappa B/metabolism , Signal Transduction/drug effects
11.
Mucosal Immunol ; 12(3): 840, 2019 05.
Article in English | MEDLINE | ID: mdl-30796335

ABSTRACT

The original version of this article contained an error in the published figures, where they appeared in black and white. These have now been corrected to display in colour.

12.
Mucosal Immunol ; 12(1): 1-9, 2019 01.
Article in English | MEDLINE | ID: mdl-29988120

ABSTRACT

The communities of bacteria that reside in the intestinal tract are in constant competition within this dynamic and densely colonized environment. At homeostasis, the equilibrium that exists between these species and strains is shaped by their metabolism and also by pathways of active antagonism, which drive competition with related and unrelated strains. Importantly, these normal activities contribute to colonization resistance by the healthy microbiota, which includes the ability to prevent the expansion of potential pathogens. Disruption of the microbiota, resulting from, for example, inflammation or antibiotic use, can reduce colonization resistance. Pathogens that engraft following disruption of the microbiota are often adapted to expand into newly created niches and compete in an altered gut environment. In this review, we examine both the interbacterial mechanisms of colonization resistance and the strategies of pathogenic strains to exploit gaps in colonization resistance.


Subject(s)
Bacteria , Bacterial Infections/immunology , Bacterial Physiological Phenomena , Dysbiosis/immunology , Inflammation/immunology , Intestines/microbiology , Microbiota , Animals , Bacterial Infections/microbiology , Dysbiosis/microbiology , Homeostasis , Humans , Inflammation/microbiology , Intestines/immunology
13.
J Exp Med ; 216(1): 84-98, 2019 01 07.
Article in English | MEDLINE | ID: mdl-30563917

ABSTRACT

Klebsiella pneumoniae, Escherichia coli, and other members of the Enterobacteriaceae family are common human pathogens that have acquired broad antibiotic resistance, rendering infection by some strains virtually untreatable. Enterobacteriaceae are intestinal residents, but generally represent <1% of the adult colonic microbiota. Antibiotic-mediated destruction of the microbiota enables Enterobacteriaceae to expand to high densities in the colon, markedly increasing the risk of bloodstream invasion, sepsis, and death. Here, we demonstrate that an antibiotic-naive microbiota suppresses growth of antibiotic-resistant clinical isolates of Klebsiella pneumoniae, Escherichia coli, and Proteus mirabilis by acidifying the proximal colon and triggering short chain fatty acid (SCFA)-mediated intracellular acidification. High concentrations of SCFAs and the acidic environment counter the competitive edge that O2 and NO3 respiration confer upon Enterobacteriaceae during expansion. Reestablishment of a microbiota that produces SCFAs enhances clearance of Klebsiella pneumoniae, Escherichia coli, and Proteus mirabilis from the intestinal lumen and represents a potential therapeutic approach to enhance clearance of antibiotic-resistant pathogens.


Subject(s)
Colon/metabolism , Drug Resistance, Bacterial , Enterobacteriaceae Infections/metabolism , Enterobacteriaceae/growth & development , Gastrointestinal Microbiome , Animals , Colon/microbiology , Colon/pathology , Enterobacteriaceae Infections/microbiology , Enterobacteriaceae Infections/pathology , Fatty Acids/metabolism , Female , Humans , Hydrogen-Ion Concentration , Male , Mice
14.
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
15.
Article in English | MEDLINE | ID: mdl-27066460

ABSTRACT

Shigella is a Gram-negative bacterium that is responsible for shigellosis. Over the years, the study of Shigella has provided a greater understanding of how the host responds to bacterial infection, and how bacteria have evolved to effectively counter the host defenses. In this review, we provide an update on some of the most recent advances in our understanding of pivotal processes associated with Shigella infection, including the invasion into host cells, the metabolic changes that occur within the bacterium and the infected cell, cell-to-cell spread mechanisms, autophagy and membrane trafficking, inflammatory signaling and cell death. This recent progress sheds a new light into the mechanisms underlying Shigella pathogenesis, and also more generally provides deeper understanding of the complex interplay between host cells and bacterial pathogens in general.


Subject(s)
Dysentery, Bacillary/pathology , Host-Pathogen Interactions/immunology , Intestinal Mucosa/pathology , Shigella/immunology , Shigella/pathogenicity , Autophagy/immunology , Dysentery, Bacillary/microbiology , Humans , Immunity, Innate/immunology , Intestinal Mucosa/microbiology , Nod1 Signaling Adaptor Protein/immunology , Nod2 Signaling Adaptor Protein/immunology , Signal Transduction/immunology , Type III Secretion Systems/metabolism
16.
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
17.
Curr Opin Microbiol ; 23: 163-70, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25497773

ABSTRACT

The role of autophagy in the control of intracellular bacterial pathogens, also known as xenophagy, is well documented. Here, we highlight recent advances in the field of xenophagy. We review the importance of bacterial targeting by ubiquitination, diacylglycerol (DAG) or proteins such as Nod1, Nod2, NDP52, p62, NBR1, optineurin, LRSAM1 and parkin in the process of xenophagy. The importance of metabolic sensors, such as mTOR and AMPK, in xenophagy induction is also discussed. We also review the in vitro and in vivo evidence that demonstrate a global role for xenophagy in the control of bacterial growth. Finally, the mechanisms evolved by bacteria to escape xenophagy are presented.


Subject(s)
Autophagy , Bacteria/immunology , Host-Pathogen Interactions , Biomedical Research/trends , Cell Biology/trends , Immune Evasion
18.
Nat Rev Immunol ; 14(1): 9-23, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24336102

ABSTRACT

Entry of bacteria into host cells is an important virulence mechanism. Through peptidoglycan recognition, the nucleotide-binding oligomerization domain (NOD) proteins NOD1 and NOD2 enable detection of intracellular bacteria and promote their clearance through initiation of a pro-inflammatory transcriptional programme and other host defence pathways, including autophagy. Recent findings have expanded the scope of the cellular compartments monitored by NOD1 and NOD2 and have elucidated the signalling pathways that are triggered downstream of NOD activation. In vivo, NOD1 and NOD2 have complex roles, both during bacterial infection and at homeostasis. The association of alleles that encode constitutively active or constitutively inactive forms of NOD2 with different diseases highlights this complexity and indicates that a balanced level of NOD signalling is crucial for the maintenance of immune homeostasis.


Subject(s)
Inflammation/immunology , Nod Signaling Adaptor Proteins/immunology , Adaptive Immunity , Animals , Autophagy , Bacterial Infections/immunology , Humans , Immunity, Innate , Intestines/immunology , Neoplasms/immunology , Nod Signaling Adaptor Proteins/chemistry , Nod Signaling Adaptor Proteins/physiology , Peptidoglycan/immunology , Signal Transduction
19.
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].

20.
Immunity ; 39(5): 858-73, 2013 Nov 14.
Article in English | MEDLINE | ID: mdl-24238340

ABSTRACT

The peptidoglycan sensor Nod2 and the autophagy protein ATG16L1 have been linked to Crohn's disease (CD). Although Nod2 and the related sensor, Nod1, direct ATG16L1 to initiate anti-bacterial autophagy, whether ATG16L1 affects Nod-driven inflammation has not been examined. Here, we uncover an unanticipated autophagy-independent role for ATG16L1 in negatively regulating Nod-driven inflammatory responses. Knockdown of ATG16L1 expression, but not that of ATG5 or ATG9a, specifically enhanced Nod-driven cytokine production. In addition, autophagy-incompetent truncated forms of ATG16L1 regulated Nod-driven cytokine responses. Mechanistically, we demonstrated that ATG16L1 interfered with poly-ubiquitination of the Rip2 adaptor and recruitment of Rip2 into large signaling complexes. The CD-associated allele of ATG16L1 was impaired in its ability to regulate Nod-driven inflammatory responses. Overall, these results suggest that ATG16L1 is critical for Nod-dependent regulation of cytokine responses and that disruption of this Nod1- or Nod2-ATG16L1 signaling axis could contribute to the chronic inflammation associated with CD.


Subject(s)
Autophagy/physiology , Carrier Proteins/physiology , Cytokines/biosynthesis , Nod1 Signaling Adaptor Protein/physiology , Nod2 Signaling Adaptor Protein/physiology , Animals , Autophagy-Related Protein 5 , Autophagy-Related Proteins , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cell Line , Crohn Disease/genetics , Crohn Disease/immunology , Crohn Disease/pathology , Cytokines/genetics , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Gene Expression Regulation , Gene Knockdown Techniques , Genetic Predisposition to Disease , Humans , Inflammation , Intestinal Mucosa/cytology , Mice , Microtubule-Associated Proteins/deficiency , Microtubule-Associated Proteins/physiology , Protein Processing, Post-Translational , RNA Interference , RNA, Small Interfering/pharmacology , Receptor-Interacting Protein Serine-Threonine Kinase 2 , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL
...