Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS Biol ; 22(1): e3002486, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38236896

ABSTRACT

Acute gastrointestinal infection with intracellular pathogens like Salmonella Typhimurium triggers the release of the proinflammatory cytokine interleukin 1ß (IL-1ß). However, the role of IL-1ß in intestinal defense against Salmonella remains unclear. Here, we show that IL-1ß production is detrimental during Salmonella infection. Mice lacking IL-1ß (IL-1ß -/-) failed to recruit neutrophils to the gut during infection, which reduced tissue damage and prevented depletion of short-chain fatty acid (SCFA)-producing commensals. Changes in epithelial cell metabolism that typically support pathogen expansion, such as switching energy production from fatty acid oxidation to fermentation, were absent in infected IL-1ß -/- mice which inhibited Salmonella expansion. Additionally, we found that IL-1ß induces expression of complement anaphylatoxins and suppresses the complement-inactivator carboxypeptidase N (CPN1). Disrupting this process via IL-1ß loss prevented mortality in Salmonella-infected IL-1ß -/- mice. Finally, we found that IL-1ß expression correlates with expression of the complement receptor in patients suffering from sepsis, but not uninfected patients and healthy individuals. Thus, Salmonella exploits IL-1ß signaling to outcompete commensal microbes and establish gut colonization. Moreover, our findings identify the intersection of IL-1ß signaling and the complement system as key host factors involved in controlling mortality during invasive Salmonellosis.


Subject(s)
Interleukin-1beta , Salmonella Infections , Animals , Humans , Mice , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Neutrophils/metabolism , Salmonella Infections/metabolism , Salmonella typhimurium/metabolism , Virulence
2.
Cell Host Microbe ; 31(3): 433-446.e4, 2023 03 08.
Article in English | MEDLINE | ID: mdl-36738733

ABSTRACT

Colonic goblet cells are specialized epithelial cells that secrete mucus to physically separate the host and its microbiota, thus preventing bacterial invasion and inflammation. How goblet cells control the amount of mucus they secrete is unclear. We found that constitutive activation of autophagy in mice via Beclin 1 enables the production of a thicker and less penetrable mucus layer by reducing endoplasmic reticulum (ER) stress. Accordingly, genetically inhibiting Beclin 1-induced autophagy impairs mucus secretion, while pharmacologically alleviating ER stress results in excessive mucus production. This ER-stress-mediated regulation of mucus secretion is microbiota dependent and requires the Crohn's-disease-risk gene Nod2. Overproduction of mucus alters the gut microbiome, specifically expanding mucus-utilizing bacteria, such as Akkermansia muciniphila, and protects against chemical and microbial-driven intestinal inflammation. Thus, ER stress is a cell-intrinsic switch that limits mucus secretion, whereas autophagy maintains intestinal homeostasis by relieving ER stress.


Subject(s)
Goblet Cells , Inflammation , Animals , Mice , Beclin-1 , Mucus , Autophagy , Intestinal Mucosa/microbiology
3.
New Microbiol ; 45(3): 193-198, 2022 07.
Article in English | MEDLINE | ID: mdl-35920874

ABSTRACT

Gastrointestinal (GI) microbial populations are important in maintaining normal functioning of the GI by preventing disorders. Dysbiotic microbiota may increase the likelihood of small intestinal bacterial overgrowth (SIBO), a syndrome associated with significant morbidity. We aimed to inves- tigate the microbiota populations of patients with SIBO. Patients with symptoms of SIBO were consecutively enrolled; they underwent a SIBO hydrogen breath test and stool was collected for microbiome analysis by sequencing of the 16S rRNA. Of the 55 patients recruited, 42 (76.4%) were positive for SIBO. When visualizing the bacterial ß-di- versity, a sub-cluster of patients was identified. Further examination of these patients' records re- vealed previous treatment for Helicobacter pylori (HP). Microbiome analysis of these patients demonstrated a significant decrease in ß-diversity (p-value<0.001) compared to patients without previous HP therapy. Furthermore, ß-diversity was significantly different in this subgroup, and sev- eral bacterial taxa were differentially expressed, including one from the genus Methanobrevibacter, which was reduced in patients that previously underwent HP treatment. Our findings suggest that while symptoms associated with SIBO may cause dysbiosis, there was no differentiation in fecal microbiome composition based on SIBO diagnosis. Furthermore, our results support previous observations regarding antibiotic-altered microbiota with effects extending two and three years post-treatment.


Subject(s)
Helicobacter Infections , Helicobacter pylori , Irritable Bowel Syndrome , Microbiota , Animals , Cattle , Dysbiosis/complications , Dysbiosis/microbiology , Helicobacter Infections/complications , Helicobacter pylori/genetics , Humans , Intestine, Small/microbiology , Irritable Bowel Syndrome/etiology , Irritable Bowel Syndrome/microbiology , RNA, Ribosomal, 16S/genetics
4.
J Immunol ; 208(12): 2713-2725, 2022 06 15.
Article in English | MEDLINE | ID: mdl-35623663

ABSTRACT

The immune system matures throughout childhood to achieve full functionality in protecting our bodies against threats. The immune system has a strong reciprocal symbiosis with the host bacterial population and the two systems co-develop, shaping each other. Despite their fundamental role in health physiology, the ontogeny of these systems is poorly characterized. In this study, we investigated the development of the BCR repertoire by analyzing high-throughput sequencing of their receptors in several time points of young C57BL/6J mice. In parallel, we explored the development of the gut microbiome. We discovered that the gut IgA repertoires change from birth to adolescence, including an increase in CDR3 lengths and somatic hypermutation levels. This contrasts with the spleen IgM repertoires that remain stable and distinct from the IgA repertoires in the gut. We also discovered that large clones that germinate in the gut are initially confined to a specific gut compartment, then expand to nearby compartments and later on expand also to the spleen and remain there. Finally, we explored the associations between diversity indices of the B cell repertoires and the microbiome, as well as associations between bacterial and BCR clusters. Our results shed light on the ontogeny of the adaptive immune system and the microbiome, providing a baseline for future research.


Subject(s)
Microbiota , Animals , High-Throughput Nucleotide Sequencing , Immunoglobulin A/genetics , Mice , Mice, Inbred C57BL , Receptors, Antigen, B-Cell/genetics
5.
Science ; 371(6529): 602-609, 2021 02 05.
Article in English | MEDLINE | ID: mdl-33303685

ABSTRACT

The gut microbiome has been shown to influence the response of tumors to anti-PD-1 (programmed cell death-1) immunotherapy in preclinical mouse models and observational patient cohorts. However, modulation of gut microbiota in cancer patients has not been investigated in clinical trials. In this study, we performed a phase 1 clinical trial to assess the safety and feasibility of fecal microbiota transplantation (FMT) and reinduction of anti-PD-1 immunotherapy in 10 patients with anti-PD-1-refractory metastatic melanoma. We observed clinical responses in three patients, including two partial responses and one complete response. Notably, treatment with FMT was associated with favorable changes in immune cell infiltrates and gene expression profiles in both the gut lamina propria and the tumor microenvironment. These early findings have implications for modulating the gut microbiota in cancer treatment.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Fecal Microbiota Transplantation/adverse effects , Gastrointestinal Microbiome , Melanoma/therapy , Nivolumab/therapeutic use , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Skin Neoplasms/therapy , Adult , CD8-Positive T-Lymphocytes/immunology , Female , Humans , Immunotherapy , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Lymphocytes, Tumor-Infiltrating/immunology , Male , Middle Aged , Programmed Cell Death 1 Receptor/immunology , Transcriptome , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
6.
JCI Insight ; 5(21)2020 11 05.
Article in English | MEDLINE | ID: mdl-33001863

ABSTRACT

Symbiotic microbial colonization through the establishment of the intestinal microbiome is critical to many intestinal functions, including nutrient metabolism, intestinal barrier integrity, and immune regulation. Recent studies suggest that education of intestinal immunity may be ongoing in utero. However, the drivers of this process are unknown. The microbiome and its byproducts are one potential source. Whether a fetal intestinal microbiome exists is controversial, and whether microbially derived metabolites are present in utero is unknown. Here, we aimed to determine whether bacterial DNA and microbially derived metabolites can be detected in second trimester human intestinal samples. Although we were unable to amplify bacterial DNA from fetal intestines, we report a fetal metabolomic intestinal profile with an abundance of bacterially derived and host-derived metabolites commonly produced in response to microbiota. Though we did not directly assess their source and function, we hypothesize that these microbial-associated metabolites either come from the maternal microbiome and are vertically transmitted to the fetus to prime the fetal immune system and prepare the gastrointestinal tract for postnatal microbial encounters or are produced locally by bacteria that were below our detection threshold.


Subject(s)
Bacteria/metabolism , Fetus/metabolism , Gastrointestinal Microbiome , Gastrointestinal Tract/metabolism , Intestines/physiology , Metabolome , Adolescent , Bacteria/genetics , Bacteria/isolation & purification , Child , DNA, Bacterial/analysis , DNA, Bacterial/genetics , Female , Fetus/microbiology , Gastrointestinal Tract/microbiology , Gestational Age , Humans , Infant , Infant, Newborn , Intestines/microbiology , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...