Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
J Neuroinflammation ; 21(1): 168, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38961498

ABSTRACT

BACKGROUND: The eye is a highly specialized sensory organ which encompasses the retina as a part of the central nervous system, but also non-neural compartments such as the transparent vitreous body ensuring stability of the eye globe and a clear optical axis. Hyalocytes are the tissue-resident macrophages of the vitreous body and are considered to play pivotal roles in health and diseases of the vitreoretinal interface, such as proliferative vitreoretinopathy or diabetic retinopathy. However, in contrast to other ocular macrophages, their embryonic origin as well as the extent to which these myeloid cells might be replenished by circulating monocytes remains elusive. RESULTS: In this study, we combine transgenic reporter mice, embryonic and adult fate mapping approaches as well as parabiosis experiments with multicolor immunofluorescence labeling and confocal laser-scanning microscopy to comprehensively characterize the murine hyalocyte population throughout development and in adulthood. We found that murine hyalocytes express numerous well-known myeloid cell markers, but concomitantly display a distinct immunophenotype that sets them apart from retinal microglia. Embryonic pulse labeling revealed a yolk sac-derived origin of murine hyalocytes, whose precursors seed the developing eye prenatally. Finally, postnatal labeling and parabiosis established the longevity of hyalocytes which rely on Colony Stimulating Factor 1 Receptor (CSF1R) signaling for their maintenance, independent of blood-derived monocytes. CONCLUSION: Our study identifies hyalocytes as long-living progeny of the yolk sac hematopoiesis and highlights their role as integral members of the innate immune system of the eye. As a consequence of their longevity, immunosenescence processes may culminate in hyalocyte dysfunction, thereby contributing to the development of vitreoretinal diseases. Therefore, myeloid cell-targeted therapies that convey their effects through the modification of hyalocyte properties may represent an interesting approach to alleviate the burden imposed by diseases of the vitreoretinal interface.


Subject(s)
Macrophages , Mice, Transgenic , Vitreous Body , Yolk Sac , Animals , Mice , Vitreous Body/cytology , Yolk Sac/cytology , Macrophages/metabolism , Mice, Inbred C57BL , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Animals, Newborn
2.
Trends Mol Med ; 30(3): 239-251, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38320941

ABSTRACT

The intestinal epithelium fulfills important physiological functions and forms a physical barrier to the intestinal lumen. Barrier function is regulated by several pathways, and its impairment contributes to the pathogenesis of inflammatory bowel disease (IBD), a chronic inflammatory condition affecting more than seven million people worldwide. Current treatment options specifically target inflammatory mediators and have led to improvement of clinical outcomes; however, a significant proportion of patients experience treatment failure. Pro-repair effects of inflammatory mediators on the epithelium are emerging. In this review we summarize current knowledge on involved epithelial pathways, identify open questions, and put recent findings into clinical perspective, and pro-repair effects. A detailed understanding of epithelial pathways integrating mucosal stimuli in homeostasis and inflammation is crucial for the development of novel, more targeted therapies.


Subject(s)
Inflammation , Inflammatory Bowel Diseases , Humans , Inflammation/pathology , Intestines , Intestinal Mucosa , Homeostasis , Inflammation Mediators/metabolism , Phenotype
4.
PLoS Pathog ; 19(6): e1011235, 2023 06.
Article in English | MEDLINE | ID: mdl-37384776

ABSTRACT

Recruitment of neutrophils into and across the gut mucosa is a cardinal feature of intestinal inflammation in response to enteric infections. Previous work using the model pathogen Salmonella enterica serovar Typhimurium (S.Tm) established that invasion of intestinal epithelial cells by S.Tm leads to recruitment of neutrophils into the gut lumen, where they can reduce pathogen loads transiently. Notably, a fraction of the pathogen population can survive this defense, re-grow to high density, and continue triggering enteropathy. However, the functions of intraluminal neutrophils in the defense against enteric pathogens and their effects on preventing or aggravating epithelial damage are still not fully understood. Here, we address this question via neutrophil depletion in different mouse models of Salmonella colitis, which differ in their degree of enteropathy. In an antibiotic pretreated mouse model, neutrophil depletion by an anti-Ly6G antibody exacerbated epithelial damage. This could be linked to compromised neutrophil-mediated elimination and reduced physical blocking of the gut-luminal S.Tm population, such that the pathogen density remained high near the epithelial surface throughout the infection. Control infections with a ssaV mutant and gentamicin-mediated elimination of gut-luminal pathogens further supported that neutrophils are protecting the luminal surface of the gut epithelium. Neutrophil depletion in germ-free and gnotobiotic mice hinted that the microbiota can modulate the infection kinetics and ameliorate epithelium-disruptive enteropathy even in the absence of neutrophil-protection. Together, our data indicate that the well-known protective effect of the microbiota is augmented by intraluminal neutrophils. After antibiotic-mediated microbiota disruption, neutrophils are central for maintaining epithelial barrier integrity during acute Salmonella-induced gut inflammation, by limiting the sustained pathogen assault on the epithelium in a critical window of the infection.


Subject(s)
Neutrophils , Salmonella Infections , Animals , Mice , Salmonella typhimurium , Epithelial Cells , Anti-Bacterial Agents , Inflammation , Epithelium , Intestinal Mucosa
5.
Mucosal Immunol ; 15(4): 656-667, 2022 04.
Article in English | MEDLINE | ID: mdl-35589985

ABSTRACT

Nuclear factor-κB (NF-κB) is a transcription factor with a key role in a great variety of cellular processes from embryonic development to immunity, the outcome of which depends on the fine-tuning of NF-κB activity. The development of sensitive and faithful reporter systems to accurately monitor the activation status of this transcription factor is therefore desirable. To address this need, over the years a number of different approaches have been used to generate NF-κB reporter mice, which can be broadly subdivided into bioluminescence- and fluorescence-based systems. While the former enables whole-body visualization of the activation status of NF-κB, the latter have the potential to allow the analysis of NF-κB activity at single-cell level. However, fluorescence-based reporters frequently show poor sensitivity and excessive background or are incompatible with high-throughput flow cytometric analysis. In this work we describe the generation and analysis of ROSA26 knock-in NF-κB reporter (KappaBle) mice containing a destabilized EGFP, which showed sensitive, dynamic, and faithful monitoring of NF-κB transcriptional activity at the single-cell level of various cell types during inflammatory and infectious diseases.


Subject(s)
NF-kappa B , Transcription Factors , Animals , Gene Expression Regulation , Genes, Reporter , Green Fluorescent Proteins/genetics , Mice , NF-kappa B/metabolism , Transcription Factors/metabolism
6.
Front Immunol ; 13: 822867, 2022.
Article in English | MEDLINE | ID: mdl-35237268

ABSTRACT

The interests in intestinal epithelial tuft cells, their basic physiology, involvement in immune responses and relevance for gut diseases, have increased dramatically over the last fifteen years. A key discovery in 2016 of their close connection to helminthic and protozoan infection has further spurred the exploration of these rare chemosensory epithelial cells. Although very sparse in number, tuft cells are now known as important sentinels in the gastrointestinal tract as they monitor intestinal content using succinate as well as sweet and bitter taste receptors. Upon stimulation, tuft cells secrete a broad palette of effector molecules, including interleukin-25, prostaglandin E2 and D2, cysteinyl leukotriene C4, acetylcholine, thymic stromal lymphopoietin, and ß-endorphins, some of which with immunomodulatory functions. Tuft cells have proven indispensable in anti-helminthic and anti-protozoan immunity. Most studies on tuft cells are based on murine experiments using double cortin-like kinase 1 (DCLK1) as a marker, while human intestinal tuft cells can be identified by their expression of the cyclooxygenase-1 enzyme. So far, only few studies have examined tuft cells in humans and their relation to gut disease. Here, we present an updated view on intestinal epithelial tuft cells, their physiology, immunological hub function, and their involvement in human disease. We close with a discussion on how tuft cells may have potential therapeutic value in a clinical context.


Subject(s)
Epithelial Cells , Helminths , Intestinal Diseases , Animals , Doublecortin-Like Kinases , Epithelial Cells/pathology , Humans , Intestinal Diseases/metabolism , Intestinal Mucosa/pathology , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Protein Serine-Threonine Kinases
7.
Blood Adv ; 5(23): 5002-5015, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34581809

ABSTRACT

Hematopoiesis is maintained by hematopoietic stem and progenitor cells that are located in the bone marrow (BM) where they are embedded within a complex supportive microenvironment consisting of a multitude of various non-hematopoietic and hematopoietic cell types. The BM microenvironment not only regulates steady-state hematopoiesis by provision of growth factors, cytokines, and cell-cell contact but is also an emerging key player during the adaptation to infectious and inflammatory insults (emergency hematopoiesis). Through a combination of gene expression analyses in prospectively isolated non-hematopoietic BM cell populations and various mouse models, we found that BM CXCL12-abundant reticular (CAR) cells are a major source of systemic and local BM interleukin-6 (IL-6) levels during emergency hematopoiesis after lipopolysaccharide (LPS) stimulation. Importantly, although IL-6 is dispensable during the initial phase of LPS-induced emergency hematopoiesis, it is required to sustain an adequate hematopoietic output during chronic repetitive inflammation. Our data highlight the essential role of the non-hematopoietic BM microenvironment for the sensing and integration of pathogen-derived signals into sustained demand-adapted hematopoietic responses.


Subject(s)
Interleukin-6 , Lipopolysaccharides , Animals , Bone Marrow , Hematopoiesis , Hematopoietic Stem Cells , Interleukin-6/genetics , Lipopolysaccharides/pharmacology , Mice
8.
J Exp Med ; 218(11)2021 11 01.
Article in English | MEDLINE | ID: mdl-34529751

ABSTRACT

Intestinal epithelial cell (IEC) NF-κB signaling regulates the balance between mucosal homeostasis and inflammation. It is not fully understood which signals tune this balance and how bacterial exposure elicits the process. Pure LPS induces epithelial NF-κB activation in vivo. However, we found that in mice, IECs do not respond directly to LPS. Instead, tissue-resident lamina propria intercrypt macrophages sense LPS via TLR4 and rapidly secrete TNF to elicit epithelial NF-κB signaling in their immediate neighborhood. This response pattern is relevant also during oral enteropathogen infection. The macrophage-TNF-IEC axis avoids responses to luminal microbiota LPS but enables crypt- or tissue-scale epithelial NF-κB responses in proportion to the microbial threat. Thereby, intercrypt macrophages fulfill important sentinel functions as first responders to Gram-negative microbes breaching the epithelial barrier. The tunability of this crypt response allows the induction of defense mechanisms at an appropriate scale according to the localization and intensity of microbial triggers.


Subject(s)
Anti-Bacterial Agents/metabolism , Epithelial Cells/metabolism , Intestinal Mucosa/metabolism , Intestines/metabolism , NF-kappa B/metabolism , Tumor Necrosis Factors/metabolism , Animals , Gene Expression Regulation/physiology , Inflammation/metabolism , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Signal Transduction/physiology
9.
Mucosal Immunol ; 14(3): 615-629, 2021 05.
Article in English | MEDLINE | ID: mdl-33731826

ABSTRACT

The gut epithelium is a critical protective barrier. Its NAIP/NLRC4 inflammasome senses infection by Gram-negative bacteria, including Salmonella Typhimurium (S.Tm) and promotes expulsion of infected enterocytes. During the first ~12-24 h, this reduces mucosal S.Tm loads at the price of moderate enteropathy. It remained unknown how this NAIP/NLRC4-dependent tradeoff would develop during subsequent infection stages. In NAIP/NLRC4-deficient mice, S.Tm elicited severe enteropathy within 72 h, characterized by elevated mucosal TNF (>20 pg/mg) production from bone marrow-derived cells, reduced regeneration, excessive enterocyte loss, and a collapse of the epithelial barrier. TNF-depleting antibodies prevented this destructive pathology. In hosts proficient for epithelial NAIP/NLRC4, a heterogeneous enterocyte death response with both apoptotic and pyroptotic features kept S.Tm loads persistently in check, thereby preventing this dire outcome altogether. Our results demonstrate that immediate and selective removal of infected enterocytes, by locally acting epithelium-autonomous NAIP/NLRC4, is required to avoid a TNF-driven inflammatory hyper-reaction that otherwise destroys the epithelial barrier.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Calcium-Binding Proteins/metabolism , Enterocytes/immunology , Inflammation/immunology , Intestinal Mucosa/pathology , Neuronal Apoptosis-Inhibitory Protein/metabolism , Salmonella Infections/immunology , Salmonella typhimurium/physiology , Animals , Apoptosis Regulatory Proteins/genetics , Calcium-Binding Proteins/genetics , Cells, Cultured , Cytotoxicity, Immunologic , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuronal Apoptosis-Inhibitory Protein/genetics , Tight Junctions/metabolism , Tumor Necrosis Factor-alpha/metabolism
10.
EMBO J ; 40(6): e105123, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33555074

ABSTRACT

Similar to the brain, the eye is considered an immune-privileged organ where tissue-resident macrophages provide the major immune cell constituents. However, little is known about spatially restricted macrophage subsets within different eye compartments with regard to their origin, function, and fate during health and disease. Here, we combined single-cell analysis, fate mapping, parabiosis, and computational modeling to comprehensively examine myeloid subsets in distinct parts of the eye during homeostasis. This approach allowed us to identify myeloid subsets displaying diverse transcriptional states. During choroidal neovascularization, a typical hallmark of neovascular age-related macular degeneration (AMD), we recognized disease-specific macrophage subpopulations with distinct molecular signatures. Our results highlight the heterogeneity of myeloid subsets and their dynamics in the eye that provide new insights into the innate immune system in this organ which may offer new therapeutic targets for ophthalmological diseases.


Subject(s)
Choroid/blood supply , Eye/immunology , Macrophages/immunology , Myeloid Cells/immunology , Neovascularization, Physiologic/physiology , Animals , Choroid/embryology , Computational Biology , Computer Simulation , Eye/cytology , Eye/metabolism , Female , Homeostasis/immunology , Humans , Immunity, Innate/immunology , Macular Degeneration/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/physiology , Myeloid Cells/metabolism , Sequence Analysis, RNA , Single-Cell Analysis , Transcription, Genetic/genetics
11.
Immunology ; 162(4): 341-356, 2021 04.
Article in English | MEDLINE | ID: mdl-32931019

ABSTRACT

Host-microbe interactions are highly dynamic in space and time, in particular in the case of infections. Pathogen population sizes, microbial phenotypes and the nature of the host responses often change dramatically over time. These features pose particular challenges when deciphering the underlying mechanisms of these interactions experimentally, as traditional microbiological and immunological methods mostly provide snapshots of population sizes or sparse time series. Recent approaches - combining experiments using neutral genetic tags with stochastic population dynamic models - allow more precise quantification of biologically relevant parameters that govern the interaction between microbe and host cell populations. This is accomplished by exploiting the patterns of change of tag composition in the microbe or host cell population under study. These models can be used to predict the effects of immunodeficiencies or therapies (e.g. antibiotic treatment) on populations and thereby generate hypotheses and refine experimental designs. In this review, we present tools to study population dynamics in vivo using genetic tags, explain examples for their implementation and briefly discuss future applications.


Subject(s)
Host Microbial Interactions , Models, Statistical , Population Dynamics , Animals , Humans , Immunity , Microbiota , Sequence Tagged Sites
13.
Nat Microbiol ; 5(9): 1119-1133, 2020 09.
Article in English | MEDLINE | ID: mdl-32514074

ABSTRACT

The interplay between host and pathogen relies heavily on rapid protein synthesis and accurate protein targeting to ensure pathogen destruction. To gain insight into this dynamic interface, we combined Click chemistry with pulsed stable isotope labelling of amino acids in cell culture to quantify the host proteome response during macrophage infection with the intracellular bacterial pathogen Salmonella enterica Typhimurium. We monitored newly synthesized proteins across different host cell compartments and infection stages. Within this rich resource, we detected aberrant trafficking of lysosomal proteases to the extracellular space and the nucleus. We verified that active cathepsins re-traffic to the nucleus and that these are linked to cell death. Pharmacological cathepsin inhibition and nuclear targeting of a cellular cathepsin inhibitor (stefin B) suppressed S. enterica Typhimurium-induced cell death. We demonstrate that cathepsin activity is required for pyroptotic cell death via the non-canonical inflammasome, and that lipopolysaccharide transfection into the host cytoplasm is sufficient to trigger active cathepsin accumulation in the host nucleus and cathepsin-dependent cell death. Finally, cathepsin inhibition reduced gasdermin D expression, thus revealing an unexpected role for cathepsin activity in non-canonical inflammasome regulation. Overall, our study illustrates how resolution of host proteome dynamics during infection can drive the discovery of biological mechanisms at the host-microbe interface.


Subject(s)
Cathepsins/metabolism , Cell Death/physiology , Macrophages/metabolism , Proteomics , Salmonella Infections/metabolism , Salmonella typhimurium/metabolism , Animals , Cathepsins/drug effects , Cell Death/drug effects , Cystatin B/antagonists & inhibitors , Inflammasomes/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Lipopolysaccharides/metabolism , Lysosomes/metabolism , Macrophages/microbiology , Mice , Peptide Hydrolases/metabolism , Phosphate-Binding Proteins/metabolism , Proteome , RAW 264.7 Cells , Salmonella Infections/microbiology
14.
Cell Microbiol ; 22(6): e13191, 2020 06.
Article in English | MEDLINE | ID: mdl-32068945

ABSTRACT

Intestinal epithelial organoids established from gut tissue have become a widely used research tool. However, it remains unclear how environmental cues, divergent microbiota composition and other sources of variation before, during and after establishment confound organoid properties, and how these properties relate to the original tissue. While environmental influences cannot be easily addressed in human organoids, mice offer a controlled assay-system. Here, we probed the effect of donor microbiota differences, previously identified as a confounding factor in murine in vivo studies, on organoids. We analysed the proteomes and transcriptomes of primary organoid cultures established from two colonised and one germ-free mouse colony of C57BL/6J genetic background, and compared them to their tissue of origin and commonly used cell lines. While an imprint of microbiota-exposure was observed on the proteome of epithelial samples, the long-term global impact of donor microbiota on organoid expression patterns was negligible. Instead, stochastic culture-to-culture differences accounted for a moderate variability between independently established organoids. Integration of transcriptome and proteome datasets revealed an organoid-typic expression signature comprising 14 transcripts and 10 proteins that distinguished organoids across all donors from murine epithelial cell lines and fibroblasts and closely mimicked expression patterns in the gut epithelium. This included the inflammasome components ASC, Naip1-6, Nlrc4 and Caspase-1, which were highly expressed in all organoids compared to the reference cell line m-ICc12 or mouse embryonic fibroblasts. Taken together, these results reveal that the donor microbiota has little effect on the organoid phenotype and suggest that organoids represent a more suitable culture model than immortalised cell lines, in particular for studies of intestinal epithelial inflammasomes.


Subject(s)
Intestine, Small/metabolism , Intestine, Small/microbiology , Organoids/metabolism , Phenotype , Proteome/metabolism , Transcriptome , Animals , Cell Line , Epithelial Cells/metabolism , Gene Expression , Humans , Inflammasomes , Male , Mice , Mice, Inbred C57BL , Microbiota
15.
Mucosal Immunol ; 13(3): 530-544, 2020 05.
Article in English | MEDLINE | ID: mdl-31953493

ABSTRACT

Inflammasomes can prevent systemic dissemination of enteropathogenic bacteria. As adapted pathogens including Salmonella Typhimurium (S. Tm) have evolved evasion strategies, it has remained unclear when and where inflammasomes restrict their dissemination. Bacterial population dynamics establish that the NAIP/NLRC4 inflammasome specifically restricts S. Tm migration from the gut to draining lymph nodes. This is solely attributable to NAIP/NLRC4 within intestinal epithelial cells (IECs), while S. Tm evades restriction by phagocyte NAIP/NLRC4. NLRP3 and Caspase-11 also fail to restrict S. Tm mucosa traversal, migration to lymph nodes, and systemic pathogen growth. The ability of IECs (not phagocytes) to mount a NAIP/NLRC4 defense in vivo is explained by particularly high NAIP/NLRC4 expression in IECs and the necessity for epithelium-invading S. Tm to express the NAIP1-6 ligands-flagella and type-III-secretion-system-1. Imaging reveals both ligands to be promptly downregulated following IEC-traversal. These results highlight the importance of intestinal epithelial NAIP/NLRC4 in blocking bacterial dissemination in vivo, and explain why this constitutes a uniquely evasion-proof defense against the adapted enteropathogen S. Tm.


Subject(s)
CARD Signaling Adaptor Proteins/metabolism , Calcium-Binding Proteins/metabolism , Neuronal Apoptosis-Inhibitory Protein/metabolism , Pathogen-Associated Molecular Pattern Molecules , Salmonella Infections/immunology , Salmonella Infections/microbiology , Salmonella typhimurium/genetics , Salmonella typhimurium/immunology , Animals , Caspases/metabolism , Disease Models, Animal , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Lymphoid Tissue/immunology , Lymphoid Tissue/metabolism , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Organ Specificity/immunology , Phagocytes/immunology , Phagocytes/metabolism , Salmonella Infections/metabolism
16.
Nat Microbiol ; 4(12): 2164-2174, 2019 12.
Article in English | MEDLINE | ID: mdl-31591555

ABSTRACT

The microbiota confers colonization resistance, which blocks Salmonella gut colonization1. As diet affects microbiota composition, we studied whether food composition shifts enhance susceptibility to infection. Shifting mice to diets with reduced fibre or elevated fat content for 24 h boosted Salmonella Typhimurium or Escherichia coli gut colonization and plasmid transfer. Here, we studied the effect of dietary fat. Colonization resistance was restored within 48 h of return to maintenance diet. Salmonella gut colonization was also boosted by two oral doses of oleic acid or bile salts. These pathogen blooms required Salmonella's AcrAB/TolC-dependent bile resistance. Our data indicate that fat-elicited bile promoted Salmonella gut colonization. Both E. coli and Salmonella show much higher bile resistance than the microbiota. Correspondingly, competitive E. coli can be protective in the fat-challenged gut. Diet shifts and fat-elicited bile promote S. Typhimurium gut infections in mice lacking E. coli in their microbiota. This mouse model may be useful for studying pathogen-microbiota-host interactions, the protective effect of E. coli, to analyse the spread of resistance plasmids and assess the impact of food components on the infection process.


Subject(s)
Dietary Fats/administration & dosage , Escherichia coli/physiology , Gastrointestinal Microbiome , Microbial Interactions , Salmonella typhimurium/physiology , Animal Feed , Animals , Bile Acids and Salts/administration & dosage , Female , Host-Pathogen Interactions , Male , Mice , Mice, Inbred C57BL , Oleic Acids/administration & dosage
17.
Nature ; 573(7773): 276-280, 2019 09.
Article in English | MEDLINE | ID: mdl-31485077

ABSTRACT

The emergence of antibiotic-resistant bacteria through mutations or the acquisition of genetic material such as resistance plasmids represents a major public health issue1,2. Persisters are subpopulations of bacteria that survive antibiotics by reversibly adapting their physiology3-10, and can promote the emergence of antibiotic-resistant mutants11. We investigated whether persisters can also promote the spread of resistance plasmids. In contrast to mutations, the transfer of resistance plasmids requires the co-occurrence of both a donor and a recipient bacterial strain. For our experiments, we chose the facultative intracellular entero-pathogen Salmonella enterica serovar Typhimurium (S. Typhimurium) and Escherichia coli, a common member of the microbiota12. S. Typhimurium forms persisters that survive antibiotic therapy in several host tissues. Here we show that tissue-associated S. Typhimurium persisters represent long-lived reservoirs of plasmid donors or recipients. The formation of reservoirs of S. Typhimurium persisters requires Salmonella pathogenicity island (SPI)-1 and/or SPI-2 in gut-associated tissues, or SPI-2 at systemic sites. The re-seeding of these persister bacteria into the gut lumen enables the co-occurrence of donors with gut-resident recipients, and thereby favours plasmid transfer between various strains of Enterobacteriaceae. We observe up to 99% transconjugants within two to three days of re-seeding. Mathematical modelling shows that rare re-seeding events may suffice for a high frequency of conjugation. Vaccination reduces the formation of reservoirs of persisters after oral infection with S. Typhimurium, as well as subsequent plasmid transfer. We conclude that-even without selection for plasmid-encoded resistance genes-small reservoirs of pathogen persisters can foster the spread of promiscuous resistance plasmids in the gut.


Subject(s)
Drug Resistance, Bacterial/genetics , Escherichia coli/genetics , Gastrointestinal Microbiome/genetics , Gene Transfer, Horizontal , Intestinal Mucosa/microbiology , Plasmids/genetics , Salmonella typhimurium/genetics , Animals , Anti-Bacterial Agents/pharmacology , Escherichia coli/drug effects , Feces/microbiology , Gastrointestinal Microbiome/drug effects , Mice , Models, Theoretical , Salmonella typhimurium/drug effects , Vaccination
18.
Sci Immunol ; 4(34)2019 04 12.
Article in English | MEDLINE | ID: mdl-30979796

ABSTRACT

Asymmetric partitioning of fate determinants is a mechanism that contributes to T cell differentiation. However, it remains unclear whether the ability of T cells to divide asymmetrically is influenced by their differentiation state, as well as whether enforcing asymmetric cell division (ACD) rates would have an impact on T cell differentiation and memory formation. Using the murine LCMV infection model, we established a correlation between cell stemness and the ability of CD8+ T cells to undergo ACD. Transient mTOR inhibition was proven to increase ACD rates in naïve and memory cells and to install this ability in exhausted CD8+ T cells. Functionally, enforced ACD correlated with increased memory potential, leading to more efficient recall response and viral control upon acute or chronic LCMV infection. Moreover, transient mTOR inhibition also increased ACD rates in human CD8+ T cells. Transcriptional profiling revealed that progenies emerging from enforced ACD exhibited more pronounced early memory signatures, which functionally endowed these cells with better survival in the absence of antigen exposure and more robust homing to secondary lymphoid organs, providing critical access to survival niches. Our data provide important insights into how ACD can improve long-term survival and function of T cells and open new perspectives for vaccination and adoptive T cell transfer therapies.


Subject(s)
Arenaviridae Infections/therapy , Asymmetric Cell Division/drug effects , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/drug effects , Immunotherapy, Adoptive/methods , Sirolimus/pharmacology , Animals , Arenaviridae Infections/immunology , Arenaviridae Infections/virology , Asymmetric Cell Division/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/physiology , Humans , Lymphocytic choriomeningitis virus/immunology , Mice , Signal Transduction/drug effects , Signal Transduction/immunology , Sirolimus/therapeutic use , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism
19.
Microbiol Spectr ; 7(2)2019 03.
Article in English | MEDLINE | ID: mdl-30953432

ABSTRACT

Bacterial infection results in a dynamic interplay between the pathogen and its host. The underlying interactions are multilayered, and the cellular responses are modulated by the local environment. The intestine is a particularly interesting tissue regarding host-pathogen interaction. It is densely colonized by commensal microbes and a portal of entry for ingested pathogens. This necessitates constant monitoring of microbial stimuli in order to maintain homeostasis during encounters with benign microbiota and to trigger immune defenses in response to bacterial pathogens. Homeostasis is maintained by physical barriers (the mucus layer and epithelium), chemical defenses (antimicrobial peptides), and innate immune responses (NLRC4 inflammasome), which keep the bacteria from reaching the sterile lamina propria. Intestinal pathogens represent potent experimental tools to probe these barriers and decipher how pathogens can circumvent them. The streptomycin mouse model of oral Salmonella enterica serovar Typhimurium infection provides a well-characterized, robust experimental system for such studies. Strikingly, each stage of the gut tissue infection poses a different set of challenges to the pathogen and requires tight control of virulence factor expression, host response modulation, and cooperation between phenotypic subpopulations. Therefore, successful infection of the intestinal tissue relies on a delicate and dynamic balance between responses of the pathogen and its host. These mechanisms can be deciphered to their full extent only in realistic in vivo infection models.


Subject(s)
Host-Pathogen Interactions/immunology , Intestinal Mucosa/microbiology , Mouth Diseases/microbiology , Salmonella Infections, Animal/immunology , Salmonella Infections, Animal/microbiology , Salmonella enterica/pathogenicity , Animals , Apoptosis Regulatory Proteins , Bacterial Proteins/genetics , Calcium-Binding Proteins , Cytokines/metabolism , Disease Models, Animal , Enterocytes/immunology , Enterocytes/microbiology , Gene Expression Regulation, Bacterial , Genes, Bacterial/genetics , Homeostasis , Immunity, Innate , Inflammation , Intestines/microbiology , Mice , Salmonella enterica/genetics , Serogroup , Streptomycin/pharmacology , Virulence Factors
20.
Cell Stem Cell ; 21(2): 225-240.e5, 2017 08 03.
Article in English | MEDLINE | ID: mdl-28736216

ABSTRACT

Bacterial infection leads to consumption of short-lived innate immune effector cells, which then need to be replenished from hematopoietic stem and progenitor cells (HSPCs). HSPCs express pattern recognition receptors, such as Toll-like receptors (TLRs), and ligation of these receptors induces HSPC mobilization, cytokine production, and myeloid differentiation. The underlying mechanisms involved in pathogen signal transduction in HSCs and the resulting biological consequences remain poorly defined. Here, we show that in vivo lipopolysaccharide (LPS) application induces proliferation of dormant HSCs directly via TLR4 and that sustained LPS exposure impairs HSC self-renewal and competitive repopulation activity. This process is mediated via TLR4-TRIF-ROS-p38, but not MyD88 signaling, and can be inhibited pharmacologically without preventing emergency granulopoiesis. Live Salmonella Typhimurium infection similarly induces proliferative stress in HSCs, in part via TLR4-TRIF signals. Thus, while direct TLR4 activation in HSCs might be beneficial for controlling systemic infection, prolonged TLR4 signaling has detrimental effects and may contribute to inflammation-associated HSPC dysfunction.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Hematopoietic Stem Cells/metabolism , Immunity, Innate , Salmonella typhimurium/physiology , Signal Transduction , Toll-Like Receptor 4/metabolism , Animals , Cell Proliferation/drug effects , Cell Self Renewal/drug effects , DNA/metabolism , Enzyme Activation/drug effects , Gene Expression Profiling , Hematopoietic Stem Cells/drug effects , Immunity, Innate/drug effects , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/metabolism , Reactive Oxygen Species/metabolism , Salmonella typhimurium/drug effects , Signal Transduction/drug effects , Stress, Physiological/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...